You are on page 1of 59

Journal of Drug Targeting

ISSN: 1061-186X (Print) 1029-2330 (Online) Journal homepage: http://www.tandfonline.com/loi/idrt20

Liposome: composition, characterization,


preparation, and recent innovation in clinical
applications

Kamel S. Ahmed, Saied A. Hussein, Abdelmoneim H. Ali, Sameh A. Korma,


Qiu Lipeng & Chen Jinghua

To cite this article: Kamel S. Ahmed, Saied A. Hussein, Abdelmoneim H. Ali, Sameh
A. Korma, Qiu Lipeng & Chen Jinghua (2018): Liposome: composition, characterization,
preparation, and recent innovation in clinical applications, Journal of Drug Targeting, DOI:
10.1080/1061186X.2018.1527337

To link to this article: https://doi.org/10.1080/1061186X.2018.1527337

Accepted author version posted online: 21


Sep 2018.

Submit your article to this journal

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=idrt20
Liposome: composition, characterization, preparation, and recent innovation in clinical

applications

Kamel S. Ahmed a,b, Saied A. Hussein c, Abdelmoneim H. Ali d, Sameh A. Korma d, Lipeng Qiu a*,

Jinghua Chena*

t
ip
a
Department of Pharmaceutics, School of Pharmaceutical Sciences, Jiangnan University, 1800 Lihu

cr
Road, Wuxi 214122, Jiangsu, PR China

us
b
Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia 11432, Egypt
c an
Biomaterial Department, College of Life Science and Technology, Huazhong University of

Science and Technology, Wuhan 430074, Hubei, PR China


M
d
State Key Laboratory of Food Science and Technology, Synergetic Innovation Center of Food

Safety and Nutrition, School of Food Science and Technology, Jiangnan University, 1800 Lihu
e d

Road, Wuxi 214122, Jiangsu, PR China


pt
ce

* Corresponding authors:

Lipeng Qiu Email: flyqlp@163.com


Ac

Tel: +86-17701518734 fax: +86-17701518734

Jinghua Chen Email: chenjinghua@jiangnan.edu.cn

Tel: + 86-51085911900 fax: +86-51085329042


Abstract

In the last decades, pharmaceutical interested researches aimed to develop novel and

innovative drug delivery techniques in the medical and pharmaceutical fields. Recently,

phospholipid vesicles (Liposomes) are the most known versatile assemblies in the drug delivery

systems. The discovery of liposomes arises from self-forming enclosed phospholipid bilayer upon

coming in contact with the aqueous solution. Liposomes are uni or multilamellar vesicles consisted

t
ip
of phospholipids produced naturally or synthetically, which are readily non-toxic, biodegradable,

and are readily produced on a large scale. Various phospholipids, for instance, soybean, egg yolk,

cr
synthetic, and hydrogenated phosphatidylcholine consider the most popular types used in different

us
kinds of formulations. This review summarizes liposomes composition, characterization, methods
an
of preparation, and their applications in different medical fields including cancer therapy, vaccine,

ocular delivery, wound healing, and some dermatological applications.


M
Keywords: Liposomes; phospholipids; applications; drug delivery; cancer; wound healing.

Abbreviations:
e d

PC: phosphatidylcholine; PE: phosphatidylethanolamine; PS: phosphatidylserine; PI:


pt

phosphatidylinositol; PA: phosphatidic acid; PG: phosphatidylglycerol; SMs: Sphingomyelins;


ce

LPC: lysophosphatidylcholine; DOPE: 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine; DOTAP:

1,2-Dioleoyl-3-trimethylammonium propane; MPER: membrane-proximal external region; DOPC:


Ac

1,2-Dioleoyl-sn-glycero-3-phosphocholine; DOPG: 1,2-dioleoyl-sn-glycero-3-phospho-(1′-rac-

glycero1; DPC: dimethyl dioctadecyl ammonium, Poly I: C and cholesterol; bFGF: basic fibroblast

growth factor; TRT: tretinoin; LSSPL: liposomes loaded with lipase-sensitive singlet oxygen-

producible (pullulan-pheophorbide a (PU-Pheo A); PUVA: Psoralen combined with ultraviolet A

radiation.
1 Introduction

Liposomes are one of the main advanced approaches in drug delivery systems. Researches

related to liposomes have gained an attractive importance in pharmaceutical, biological, and

medical fields since liposomes consider the most proper carriers for the introduction of all kinds of

agents, such as anticancer drugs [1, 2], antibiotics [3, 4], anti-inflammatory [5, 6], genes [7, 8], and

antifungal [9, 10]. Liposomes are the first enclosed microscopic phospholipid bilayer systems,

t
ip
which were approved in 1965 [11, 12]. They are circular soft-matter vesicles formed from one or

more bilayer membrane(s) separating an aqueous medium from another. Phospholipid molecules

cr
mainly composed of different polar head groups and two hydrophobic hydrocarbon chains. The

us
polar groups can be zwitter ionic or negatively charged. The hydrocarbon chain molecules have
an
different lengths and possess different degrees of unsaturation. The formation of liposomes occurs

spontaneously upon reconstitution of dry lipid films in an aqueous solution [13, 14].
M
The main advantages of systemic liposomes as drug formulations arise from their

biodegradability, lowers systemic toxicity, targeted delivery, sensitive molecules protection, and
e d

improved pharmacokinetic effects. On the other hand, their advantages for the topical applications
pt

accrue from the demonstrated ability to decrease serious incompatibilities and side effects that
ce

might originate from the undesirably large systemic drugs absorption, the significant enhancement

of drug accumulation at the required site of action due to the high similarity between the liposome
Ac

composition and the biological membranes [15].

Many types of research have been carried out for the surface modification of liposomes to

enhance their properties and increase their applicability to deliver the active pharmaceutical

ingredients to the site of action for in-vivo applications. For example, the utilization of polyethylene

glycol (PEG) and polyvinyl alcohol (PVA) decreased opsonization and made the liposomes less
susceptible for hepatic clearance, and consequently prolonged liposomes residence time in the

systemic circulation [6, 16]. As well, the insertion of rhodamine-123-conjugated polymer in the

surface of the liposomes increased the formulation accumulation in the mitochondria [17]. The

incorporation of D-α-tocopheryl polyethylene glycol 1000 succinate-triphenylphosphine conjugate

(TPGS1000-TPP) to the surface of a liposomal formulation of paclitaxel improved its cellular

uptake and mitochondrial targeting and consequently enhanced apoptosis processes in drug-

t
ip
resistant lung cancer [18]. The previously mentioned advantages and the simple handling and

modulation processes of liposomes explain their strong invasion for the scientific research area and

cr
various biological applications and also explain the abundant presence of liposome preparations in

us
market and clinical trials. there are a number of liposomal products such as ambisome, myocet,
an
doxil, depoCyt, etc., approved by the FDA for commercial usage. Table 1 showing some liposomal

formulation recently used in the global liposomes drug delivery market and some others still under
M
clinical trials.

2 Composition of liposomes
e d

Liposomes are mainly composed of phospholipids, which contain two major categories
pt

including glycerophospholipids and sphingomyelins. The phospholipids of eukaryotic cells are


ce

mainly glycerophospholipids, in which glycerol considered the backbone. The chemical structure of

glycerophospholipids consisted of a hydrophilic head group and a hydrophobic side chain. Different
Ac

glycerophospholipids are obtained as a result of head group variation, for example,

phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylserine (PS),

phosphatidylinositol (PI), phosphatidic acid (PA), phosphatidylglycerol (PG), and cardiolipin (CL)

[19, 20]. As well, the length variation of the nonpolar moieties results in various
glycerophospholipids, such as dimyristoyl, dipalmitoyl, or distearoyl PC. Furthermore, the bonding

type (ether or ester) between glycerol and aliphatic chains results in different glycerophospholipids.

Sphingomyelins (SMs) are a vital membrane component of the animal cells [21, 22].

Sphingosine is the backbone of SM; each molecule of SM averagely has cis-double bonds in amide-

linked acyl chains. The natural SMs have typical acyl lengths usually longer than the paraffin

residues of sphingosine, so they considered as asymmetric molecules. SMs have the capability to

t
ip
form intermolecular and intramolecular hydrogen bonds. SMs have been used in the liposomal

formulation and showed good entrapment efficiency, greater serum stability, with a rapid release

cr
profile in comparison with the DSPC liposomes [23]. Another study showed that a spherically

us
shaped vesicle with small particle size has been successfully prepared from sphingomyelin solution
an
by using ultrasonic–supercritical CO2 technique but the size distribution of the vesicles increased
M
by increasing the operating temperature as a result of aggregation [24]

2.1 The abundant sources of phospholipids


d

Animal tissues (bovine brain and egg yolk), and vegetable oils (soybean, corn, cotton seed,
e

sunflower, and rapeseed) are considered the common phospholipids sources. Soybean oil and egg
pt

yolk are the common important sources of phospholipids. Egg yolk is distinguished with (1) larger
ce

quantities of PC, (2) phospholipids with long-chain polyunsaturated fatty acids (arachidonic acid
Ac

and docosahexaenoic acid), (3) the presence of SM, (4) egg yolk lecithins of the higher saturation

level, which could cause better have been used for phospholipids extraction [25]. In a recent study,

ethanol and butanol with different contents of water were assessed for phospholipids extraction.

The yolk flakes were prepared by using a small drum dryer heated with saturated steam. The liquid

yolk was applied on the smooth hot surfaces of the drums, where denaturation of yolk protein and

water evaporation takes place. The dried yolk was collected as “flakes” from the surface of the
drum by a scraper, and the dried flakes were then packed into a column through which the organic

solvent from the reservoir was added to submerge the flakes, followed by solvent evaporation from

the lipid extract under vacuum by using a rotary evaporator. Then, the lipid extracts were dried at

40 °C for 5 h in a vacuum oven. All yolk lipids were effectively extracted by using butanol with a

little preference for phospholipids, while lipids extraction by using aqueous ethanol was affected by

its water content since ethanol of 75% exhibited the highest preference for phospholipids [26]. In

t
ip
another study, three solvents (butanol 100%, butanol 80%, and ethanol 95%) were used for the

extraction of phospholipids by injecting the yolk as a thin stream into the hot solvent, after that the

cr
liquid yolk was solidified upon contact with the solvent into the thread. 100 g of egg yolk was

us
vortexed and texturized in 200 mL of the solvent under similar conditions. The de-oiled yolk thread
an
was subjected to extraction 4 more times sequentially, then the solvent was evaporated using a

rotary evaporator, and the obtained lipids were dried at 40 °C for 5 h in a vacuum oven. It was
M
shown that the total yolk lipids were effectively extracted by using butanol, and the extraction was

faster than that of ethanol, but with little preference for phospholipids. Conversely, ethanol had a
e d

more preference for phospholipids than for neutral lipids [27].


pt

Liu, Zhou [28], extracted phospholipids from edible clams (Cycling Sinensis, Mactra
ce

Chinensis Philippi, Mactra veneriformis Reeve, Meretrix meretrix, Saxidomus purpurata, and

Ruditapes phliippinarum) by using a mixture of hexane and ethanol. Clam meat powder was mixed
Ac

with hexane/ethanol (1:1, v/v), and vortexed for 1 min, stirred at 50 °C for 90 min, and then the

mixture was centrifuged at 7,800 g for 10 min. Finally, the organic layer was collected and dried at

35 °C under nitrogen. Large quantities of the polyunsaturated fatty acids (docosahexaenoic acid and

eicosapentaenoic acid) were recovered. The extracted oils contained a high percentage of
phospholipids making up 39.86–74.05% of the total lipids. PC (37.40–52.19%) and PE (34.74–

43.10%) were the predominant species.

Phospholipids classes from duck, quail, and hen egg yolks were separated and identified.

Egg yolk was separated from the egg white and transferred into a 200-mL vial, followed by the

addition of ethanol, and stirring for 10 min with a magnetic stirrer. The solvent layer was filtered

into a round-bottom flask, and the residual was extracted with ethanol twice. The ethanolic solution

t
ip
was crystallized under the lower temperature at 2–5 °C for 2 h, then ethanol was evaporated from

the solution by a rotary evaporator, and the extracts were collected. The residue was then dissolved

cr
in hexane and transferred into a vial placed in an ice bath. Finally, 120 mL of cold acetone (-20 °C)

us
was carefully poured into the stirring mixture until phospholipids precipitation. The obtained results
an
revealed that 57 molecular species of egg yolk phospholipids were detected and identified. Among

the different classes of egg yolk phospholipids, PC (16:0–18:1), PE (18:0–20:4), PI (18:0–18:2), PS


M
(18:0–18:2), SM (d18:1/16:0), and lysophosphatidylcholine (LPC) (16:0) were the predominant

molecular species [29].


e d

Gas chromatography coupled with mass spectrometry (MS) and hydrophilic interaction
pt

liquid chromatography was used for the determination of omega fatty acids profile in egg yolk.
ce

Separation was accomplished by the liquid chromatography by using columns (stainless steel)

packed with a homemade alkyl-amide stationary bonded phase (synthesized on Kromasil 100 silica
Ac

gel). A diode array detector (SPD-M20A) was used for measuring the absorbance at 206 nm.

MS/MS analysis was achieved under the positive ionization mode on a triple quadruple coupled

with an electrospray ionization source. During 20 min the phospholipids were eluted in the

following order: PG, LPG, PE, LPE, PC, SM, and LPC. Furthermore, palmitic acid was the major

saturated fatty acid detected [30].


Phospholipids fractions were extracted from Camelina sativa seeds by using the

supercritical carbon dioxide method. A total phospholipid content of 360 and 130 mg/kg lipid was

obtained by the hexane-extracted and cold-pressed oils, respectively. While the total content of the

phospholipid extracted with pure supercritical carbon dioxide method ranged between 520 (45

MPa/70 °C) and 2000 mg/kg lipid (45 MPa/50 °C). PI, PE, and PC were the major classes of

phospholipids found in the lipids of camelina seeds, and PI was the predominant species. More

t
ip
phospholipids were extracted by increasing the concentration of the ethanol to 10% (w/w) [31].

3 Methods of liposomes preparation

cr
All the methods of liposomes preparation involve three to four basic steps:

us
1. Drying lipids through the evaporation of the organic solvent.

2. The lipid dispersion in an aqueous media.


an
3. Liposome purification.
M
4. Analysis of the final product.

The preparation of liposomes could be achieved by using three different techniques, such as the
e d

mechanical methods, the solvent dispersion methods, and methods based on fusion or size
pt

transformation of the prepared vesicle [32].


ce

3.1 Mechanical methods

Preparation of liposomes by the thin film hydration method


Ac

The most common simply and widely applicable method for liposomes formulation is the

thin-film hydration method [33]. In this technique, liposomes are prepared by dissolving the lipid in

an organic solvent usually chloroform or mixtures of chloroform and methanol, and the solvent is

then removed by film deposition under vacuum. After the complete evaporation of the organic

solvents, the lipid residue is hydrated by using an aqueous buffer; the lipids spontaneously undergo
swelling and hydration to form a liposome. This method produces liposomes with a heterogeneous

sized aggregation of multilamellar vesicles over one micrometer in diameter. The particles can be

downsized by using different techniques, for instance, extrusion or sonication [34, 35].

3.2 Preparation of liposomes by solvent dispersion methods

3.2.1 Ether injection method

In this method, liposomes are prepared by dissolving the lipid in diethyl ether or a mixture of

t
ip
methanol and ether, followed by the slow injection to an aqueous solution of the compound to be

cr
incorporated under lower pressure or at 55-65 °C. The main drawbacks of this procedure are the

us
heterogeneous population of the obtained vesicles, and the exposure of the substances to be

encapsulated to high temperature [36].

3.2.2 Ethanol injection method


an
M
This method involves the utilization of ethanol to dissolve phospholipids and cholesterol. By

means of a syringe pump, the resulting lipid solution is injected under stirring conditions in a
d

definite volume of the aqueous solution. Liposomes are then spontaneously formed as soon as the
e
pt

lipid solution contacts the aqueous phase. The suspension of liposomes is then left under stirring at

room temperature for 15 min. This procedure offers numerous advantages, such as fast
ce

implementation, simplicity, and reproducibility. Also, it does not lead to oxidative alterations or
Ac

degradation of lipids and has the ability to form small unilamellar vesicles without extrusion or

sonication [37, 38].


3.3 Preparation of liposomes by methods based on fusion or size transformation of prepared

the vesicle

3.3.1 Freeze-thaw extrusion method

Freezing and thawing method is considered to be a convenient technique for increasing the

trapped capacity of the liposomal preparations since it reveals a physical disruption of the lamellar

structure, likely due to the ice crystals formed during the process of freezing [39]. Liposomes

t
ip
prepared by the film method were vortexed with the material to be incorporated until the whole

lipid film is suspended, and the resulted vesicles are frozen in warm water and vortexed again. Then

cr
extrusion of the sample is applied three times after 2 cycles of freeze-thaw and vortexing, followed

us
by 6 cycles of freeze-thaw and additional 8 extrusions. Freeze-thaw cycling is a method frequently
an
used in liposomes preparation in order to increase the encapsulation efficiency [40, 41].

3.3.2 The dehydration/rehydration method


M
One of the drawbacks of macrolide and aminoglycoside antibiotics liposomes is their low
d

encapsulation efficiency, which results in preparations with lower drug content. The encapsulation
e

efficiencies and stability (in-vitro) of these antibiotics could be enhanced by


pt

dehydration/rehydration of the obtained vesicles [42]. Emptying the buffer containing small
ce

unilamellar vesicles and rehydrating it with the aqueous solution containing the compound to be

incorporated after which they are dried. This lead to solid lipids dispersion in small subdivided
Ac

forms and the vesicles are then rehydrated. Liposomes prepared by this technique are usually

oligolamellar vesicle [43, 44].


3.4 Supercritical fluid technology

The supercritical state of a fluid is intermediate between that of gas and liquids. In the

pharmaceutical field, supercritical carbon dioxide (scCO2) is the most commonly used gas which

can become supercritical at its critical temperature and pressure of 31.1°C, 7.38 respectively [45].

The supercritical anti-solvent (SAS) method.

This technique is being used to produce a micronized and homogeneous dispersion of phospholipid

t
ip
materials. Cholesterol and other lipid materials dissolved in an organic solvent and placed in a glass

cr
container. CO2 gas is sprayed through capillary tubes into a high-pressure precipitation vessel and

us
as a result of a sudden change in temperature and pressure the CO2 gas transformed into a

supercritical phase. Subsequently, evaporation of the organic solvent takes place and the lipids are
an
extracted into the supercritical phase, which leads to supersaturation of the lipids in the

scCO2 phase and precipitation of the lipid materials. After that, the organic solvent is removed
M
by continuous pumping CO2 into the vessel to achieve fine lipid particles. Finally, adding the
d

aqueous solution for liposomes formation. A previous study made a comparison between the
e

conventional method (Bangham method) and the supercritical anti-solvent (SAS) method, it was
pt

reported that SAS method was more efficient and environmentally-friendly procedures to produce
ce

liposomes. Because it produced small particle size distribution and high entrapment efficiency and

considered as an environmentally-friendly technique because it enables to apply “soft” organic


Ac

solvents for example ethanol if compared with the conventional methods organic solvents

(isopropyl ether, chloroform, diethyl ether, and methanol). And also, the SAS process is performed

under lower temperature conditions unlike the conventional Bangham method [46]. Another study

used the Supercritical process for encapsulation of both eugenol (EUG) and α-lipoic acid (ALA) as

an antioxidant in liposome vesicles. It was mentioned that EUG was entrapped with an overall
efficiency of 86.3% with a particle size of 200 nm and ALA was encapsulated with a maximum EE

of 68.1% with a particle size of 230 nm and the prepared liposome exhibited a good stability for at

least 40 days [47]. The previous studies did not show a significant difference in terms of the

liposome size, entrapment efficiency and stability compared with the conventional Bangham

method for liposome preparation except SAS process lead to complete removing of the organic

solvent. To avoids the problems caused by organic solvent residues, and due to the dissolution

t
ip
properties of supercritical CO2, that could be used as an excellent substitute for the organic solvent,

an improved Supercritical reverse phase evaporation (ISPER) method was developed in which the

cr
aqueous solution with the phospholipid materials were introduced into a sealed viewing cell.

us
followed by adjustment the temperature and pressure to suitable values and then introducing the
an
CO2. After equilibration, CO2 removed, and liposomes were formed, and the liposome prepared by

this method showed improvement in entrapment efficiency of glucose using alpha-dioleoyl


M
phosphatidylcholine (DOPC) if compared by the Bangham method. moreover, the liposome

prepared by the ISPER method was highly stable for 30 days [48]. In general, by comparing the
e d

SCF technique with other conventional methods for liposome preparation the SCF has provided
pt

several advantages for example, due to the use of supercritical carbon dioxide (scCO2) (non-
ce

flammable, inert, non-toxic, and more economic), SCF was considered as a green process,

Possibility of large-scale production of liposome by SCF under the good manufacturing practice
Ac

(cGMP) conditions [49], finally liposome once formed no need for further processing (drying and

precipitation) to achieve dry powder liposomal formulations [50].


4 Liposome characterizations

4.1 Particle size

The liposomes size considers one of the vital factors that should be controlled particularly

when these formulations are intended for the parenteral application (topical, injection, and

inhalation). Numerous procedures are available for monitoring the liposomal size, for example,

t
Sonication, Extrusion, and Homogenization.

ip
Sonication

cr
us
The multilamellar vesicles (MLVs) suspension that produced by different preparation techniques

(thin film hydration method) is transferred into test tubes and subjected for sonication by tip
an
sonication that require larger sample volume (1-5 ml) and more energy or in a bath sonicator that

provide better control of the temperature, small volumes of the sample is required (1 ml), and
M
proper for preparation that in jelly form or that doesn't swell well. The pressure stress induces
d

disruption of the large and the MLVs present in the samples into small unilamellar vesicles.
e

Sonication process normally lasts for 5–10-min and this will provide small unilamellar vesicles
pt

(20-50 nm). But the different kind of the lipids will impact on the vesicles final size, for example,
ce

diacyl cationic lipids can form micelles (dioctadecylamidoglycylspermine (DOGS)), and neutral
Ac

lipid (Dioctadecyl diammonium bromide (DOBAD)), the vesicles cannot be downsized <130 nm

[51, 52].

Extrusion

Liposome extrusion occurs by pushing the large MLVs solution through filters of polycarbonate

membrane. The desired vesicles size controlled by the applied pressure, since the average size
decreases upon increasing the extrusion pressure [53], also decreasing the pore size of the filter

membrane result in reduction in size and size distribution of the liposomes, it was reported that

using filter membrane with pore size larger than 0.2 μm the size of the obtained liposomes was

smaller than the membrane pore size, but by using a membrane with a pore size smaller than 0.2

μm, the size of the produced liposomes was slightly larger than the filter pore size [54]. And also,

another study proved that filtering of the MLVs solution using a filter membrane with pore size

t
∼1 μm followed by five times repeated filtration through 0.4- and 0.2-μm pores. Then five to ten

ip
extrusions using a 100-nm membrane pore size will result in the formation of large unilamellar

cr
vesicles with size ∼110–120 nm. If the size is needed to be smaller, continuous filtration using a

us
membrane filter with 80- and 50-nm pores size is required. The vesicles treated by the extrusion
an
techniques characterized by proper homogeneity and easy to control the vesicles size distribution,

especially for vesicles of large diameters (100–500 nm) [51].


M
Homogenization
d

The downsizing of the liposomes by this technique established by the collision of the
e
pt

larger vesicles in the interaction chamber at high pressure [55]. This method is characterized by

its simple usage for large-scale production of the liposome, high capacity (10 mL to hundreds of
ce

liters), and rapid procedures. But the probability of contamination and degradation of the sample
Ac

can occur especially with very small and certain large vesicles. Normally, minimal size can be

attained by 3-5 passages through the interaction chamber, the diameter and the entrapped volume

of the vesicles decreases with increasing the cycles number and the inlet pressure [56]. For

assessing the liposome size and size distribution, numerous methods are available, for example,

size-exclusion chromatography (SEC), field-flow fractionation and static or dynamic light

scattering, microscopy techniques.


As a result of the drawbacks of the electron microscope technique (EM) (TEM, cryo-EM, freeze

fracture TEM) like complicated sample preparation (staining and drying procedures), induce

shape alteration and shrinkage, and time-consuming, so atomic force microscopy (AFM) is a

novel rapid techniques that able to assess the three-dimensional shape of the liposome surface

without sample modification with a sharp probe or tip, and doesn't cause any damaging or

alteration effects on the liposome, but liposome dispersion analysis should be immediately after

t
ip
deposition of the sample because evaporation of the aqueous medium will lead to vesicles

rearrangements. Field-flow fractionation (FFF) is a flexible elution method, provides convenient

cr
and rapid procedures for measurement and separation of liposome size. It has the ability to separate

us
a wide range (1 nm – 100 μm) of particle sizes with high resolution. In this technique Particle size
an
separation depending on hydrodynamic size basis (flow FFF) or weight basis (sedimentation FFF).

Furthermore, when joined to other detectors like light scattering, transmission electron
M
microscopy, UV-absorbance, and atomic force microscopy can offer a proper information on

vesicle properties such as size, structural parameters, shape, and sample contamination [51, 57].
e d

Size exclusion chromatography (SEC), is a standard technique used for size separation, Separation
pt

occurs by passing the sample dispersion through a column packed with a porous material that
ce

entraps small particles and excludes large particles from the internal pore volume resulting in their

lower retention on the column. This result in larger particles firstly eluted before the smaller one. It
Ac

is characterized by the following: simple and uncomplicated technique, small factors number

required to be controlled for the experiments, simple set up, and its coupling with Refractive index,

UV-Vis, or fluorescence detectors makes it able to rapid and easy method validation and

development but there are some drawbacks, it deals with a small molecular weight range of

separation and rigidity of the analysis conditions and high sensitivity of the separation method to
several parameters like ionic strength, pH, and type of charge (cation and anion) of the carrier

solution [58].

Dynamic light scattering

The intensity of light scattered by suspended particles undergoing Brownian motion (from

collisions between suspended particles and solvent molecules) can be determined as a function of

t
time by Dynamic light scattering (DLS). This technique provides several advantages such as

ip
Accurate, repeatable and reliable size analysis in its native environment, high concentration and

cr
turbid samples can be directly measured, fully automated measurement, simple set up, measurement

us
of small sizes < 1nm and MW < 1000Da, and Low sample volume (as little as 2µL) [59, 60, 61].

4.2 Encapsulation efficiency


an
Increasing the drug encapsulation efficiency (EE) will contribute to the enhancement of the drug
M
bioavailability [62]. Different methods used for measurement of the encapsulation efficiency.
d

Firstly separation of free drug from loaded one occurs by different separation techniques
e

(centrifugation and dialysis membrane), and then direct determination of encapsulated drug
pt

achieved by solubilizing or disruption of the lipid bilayer by methanol [63, 64], chloroform [65, 66]
ce

or by triton x 100 [67, 68] followed by filtration and measurement. Indirect methods achieved by
Ac

determining the quantity of unencapsulated drugs then subtract the value from the total amount of

the drug used [69]. A recent study using liquid chromatography (nanoparticles exclusion

chromatography) for rapid measurement of encapsulation efficiency without any pretreatment for

the sample through direct injection of the liposomal suspension into the column, and the free drug

eluted according to its physical properties under appropriate mobile phase conditions [70, 71].

Another study used different techniques [(solid-phase extraction (SPE), size-exclusion


chromatography (SEC), hollow fiber centrifugal ultrafiltration (HF-CF-UF) and centrifugation

ultrafiltration (CF-UF)] for assessment of the EE of the Amphotericin B (AmB) loaded liposome.

The EE by SPE was found 5–13%, SEC was about 93%, and by HF-CF-UF was approximately

99.0% and nearly 100% by CF-UF. The paper reported that the variation of EE among these

methods back to the following reasons; by using SPE, the interaction of cholesterol with the

stationary phase result in difficult elution with water, and raise the leakage probability of liposome,

t
ip
by SEC some parameters could cause liposome leakage, such as osmotic shocks, adsorption, the

pore size and the kind of columns. While for CF-UF, the main drawback prevents the

cr
unencapsulated drug to pass freely through the membrane, leading to no detection of the

us
unencapsulated drug, was the concentration polarization, HF-CF-UF could actually reflect the
an
encapsulation efficiency of the liposomes with the unentrapped AmB concentration less than 25.0

micro g/mL, but by increasing the concentration more than 25.0 micro g/mL, lead to an increase in
M
the size of AmB molecular aggregates and consequently entrapped by hollow fiber [72]. There are

different substances that have an effect on the encapsulation efficiency, for example, cholesterol,
e d

several studies proved that the encapsulation efficiency decreased by increasing the cholesterol
pt

content, and these may result from the steric hindrance and the competition between the cholesterol
ce

and hydrophobic drug molecules for space present in the phospholipids bilayer resulting in lower

encapsulation of the drug molecules [66, 73]. Another study investigated the effect of aptamer on
Ac

encapsulation efficiency. Aptamers are DNA or RNA sequences produced to display high affinity

and specificity against a broad range of targets with poor immunogenicity, high thermal stability,

and synthesis on large scale with lower costs for production, it was reported that the loading

efficiency of doxorubicin-binding aptamers was ten folds greater than conventional doxorubicin

liposomal formulations and also encapsulation efficiency of tobramycin (hydrophilic) into


liposomes was improved 6 times by applying aptamers [74]. Another study confirmed that the

presence of transmembrane salt gradient (ammonium salts, sodium salts) had a strong effect on the

EE. It was investigated that using of salt gradients means for loading of Doxorubicin result in

charging and protonation of DXR inside the liposome, and also DXR precipitation in the interior

part of the liposome when the DXR concentration exceeded the solubility. Also showed that the use

of ammonium salt-gradients (NH4+-citrate 100%, NH4+-phosphate 98%, NH4+- sulfate 95%,

t
NH4+-acetate 77%) had higher loading efficiency than sodium salt gradient (Na+- citrate 54%, Na+-

ip
phosphate 52%, Na+- sulfate 44%, Na+- acetate 16%) [75, 76].

cr
us
4.3 Liposomal Drug release

Release studies can be performed at 37 oC under sink condition using appropriate release
an
media (buffer). a dialysis membrane with specific cut-off molecular weight used in the form of
M
dialysis bag or on the end of the tube and soaked in receptor media under stirring condition. The

release medium is usually buffer with PH 7.4 and perfect sink conditions and kept at 37 oC under
d

stirring to mimic the in vivo conditions (In-Vitro Drug Release). At predetermined time intervals,
e
pt

aliquot volumes of the medium were taken out at various time intervals for concentration detection

by different analysis technique (HPLC, UV-vis spectrophotometry), and substituted with the same
ce

volume of fresh media for keeping the volume of the receptor media constant. The release of the
Ac

drug from liposome is affected by several parameters. For instance, the presence of cholesterol in

liposome formulation will influence on the release of the hydrophilic drugs as the quantity of

cholesterol increases, the release will be rapid. Conversely, the release of the hydrophobic drugs

exhibited a reduced release action [77]. Another study confirmed that the incorporation

of cholesterol into the doxorubicin liposome decreased the undesired leakage of doxorubicin at

37°C in buffered saline and also in fetal bovine serum (FBS) [78]. Moreover, the drug release can
be controlled to overcome the problems that arise from conventional liposome like rapid leakage,

slow release of the entrapped drug, the drug cannot be controlled released in targeting sites, or the

drug may be leakage before reaching the active sites and these drawbacks can be avoided by

incorporating components in liposome formulations to achieve pH [79, 80, 81], thermal [82, 83,

84], photothermal [85, 86], enzymatic [87, 88, 89], or magnetic [90, 91, 92] triggered release.

4.4 Zeta Potential

t
ip
The ZP refers to the potential difference between the electric double layer (EDL) (an adsorbed

cr
double layer developed on the surface of dispersed charged particles) of the movable particles and

us
the layer of dispersant around them at the slipping plane. The particle surface charge value reflects

the nanosuspensions stability, considering that NP-dispersions with ZP values of ±30 mV is


an
electrostatically stabilized nanosuspension [93]. For measuring the zeta potential, an electric field
M
is applied, and the particles electrophoretic mobility is measured based on the scattering of an

incident laser by the mobile particles. Many factors affecting zeta potential value, for instance, PH,
d

ionic strength, and the particles concentration [94]. The phospholipid composition (positively and
e
pt

negatively charged phospholipids) of liposomes is the main content that influences on the overall

surface charge of the liposome [20].


ce

4.5 Freeze drying


Ac

Liposomal preparations do not meet the required long-term stability standards of

pharmaceutical formulations if stored as aqueous suspensions. Over time the incorporated active

ingredients tend to leak out of the lipid bilayer structure and vesicles liable to fuse or aggregate on

storage. So, the removal of water will improve the stability and increase the shelf life of liposomal

formulations. Freeze drying is one of the most commonly used methods for drying and enhancing
the stability of several pharmaceutical products including liposomes. The drying process includes

three separate steps, begin with freezing then ice sublimation and finally desorption of unfrozen

water, however during these stages physical changes of the vesicles such as alterations in the

vesicle size and loss of the incorporated agent may occur. Fortunately, these obstacles were solved

by using sugars such as sucrose, glucose, trehalose, mannitol, and lactose that have the ability to

interact with the headgroups of the phospholipid through formation of H-bonds leading to

t
ip
depression of phase transition temperature (Tm). Consequently, phase transitions during

dehydration and rehydration will be avoided and the loss of incorporated solutions will be

cr
prohibited. Another mechanism of the lyoprotectant is the formation of viscous glassy matrix in and

us
around the liposomes preventing the fusion process and protects vesicles against rupture during
an
formation of ice crystals [95]. The influence of different cryoprotectants on the stability of liposome

preparation of 5-Fluorouracil was studied. The data showed that lyophilized cake of liposomes
M
(without cryoprotectants) was compact and hard to reconstitute, on the other hand, when using

cryoprotectants the liposome cake was fluffy and easily reconstituted. Glucose, mannitol and
e d

trehalose were used as cryoprotectants. The protective efficacy of trehalose was more than glucose
pt

and mannitol, the efficiency of the lyophilization process increased with increasing the
ce

cryoprotectant concentration [96]. Sucrose was used as cryoprotectant in liposome preparation of

vitamin C and hydrophobic drug medium-chain fatty acids (MCFAs). liposomes were dried below
Ac

−86 °C for 48 h at a sucrose/phospholipids ratio of 2:1 (w/w). The dried liposomes exhibited high

encapsulation efficiency of MCFAs and vitamin c (44.26 ± 3.34, 62.25 ± 3.43 % respectively) with

small particle size (110.4 ± 7.28) nm and high storage stability at 4 °C for 60 days [97]. The

stability properties of liposomal preparation incorporating ftorafur and vitamin A was investigated

in presence of suitable sugars, the transition temperature of liposomal dispersions was the highest
with trehalose and the liposome size was smallest (294±8nm) than that of glucose (751±12nm),

sucrose (426±9nm), and mannitol (356±4nm), moreover, the liposome suspension with 15%

trehalose exhibited the highest retention rate 60.5 and 99.2±0.1% for ftorafur and vitamin A

respectively after freeze-drying [98]. Recently published study reported that trehalose at a 5:1 sugar

to lipid ratio had the ability to maintain the permeability properties and the structural integrity of the

prednisolone sodium phosphate liposome during the freeze-drying procedure [99]. Many factors of

t
ip
the formulation should be carefully investigated to ensure the success of freeze-drying process as

the liposome composition (phospholipid type, concentration and type of conjugated polymers, type

cr
and exact concentration of cryoprotectants, interaction between cryoprotectants and liposomes,

us
surface modification of liposomes).

4.6 Stability study


an
Stability of liposome is a complex issue. It can be classified into chemical, physical and
M
biological stabilities which are strongly inter-related to each other, the physical and chemical
d

stability influence on the liposomes-shelf life (size distribution, entrapment efficiency, and
e

compound degradation), Physical stability studies of liposomes can be achieved either by visual
pt

appearance and observations through determination of color changes and sedimentation of liposome
ce

formulations or microscopic observations by using transmission electron microscopy and atomic

force microscopy (AFM) to evaluate the particle size and the three-dimensional shape of the
Ac

liposome surface, vesicle surface charges play an important role in preventing aggregation and also

optimizing the storage conditions (temperature and humidity) [100] help in extending the liposome

shelf life stability. the chemical stability of the phospholipids from which the bilayer of the

liposome is composed is very important and should be considered especially if the vesicle

composed from unsaturated phospholipids that prone to hydrolysis and oxidation. The chemical
stability studies can be achieved by studying the phospholipid composition (hydrolysis and

peroxidation) and drug degradation using liquid chromatography, thiobarbituric acid (TBA) test

(lipid oxidation), and spectroscopy. Protection of phospholipids against oxidation can be attained by

using high-quality lipid materials void from hydroperoxides and metal ions. Liposome preparation

under an inert gas such as argon or nitrogen reduces the oxidation of lipids, Storage of liposome at

low temperatures and away from light and oxygen will decrease the chance of oxidation. Moreover,

t
ip
using antioxidants, such as butylhydroxytoluene (BHT) and a-tocopherol can protect against

oxidation. phospholipids hydrolysis can completely be avoided by water removal through freeze-

cr
drying in the presence of a suitable stabilizing cryoprotectant [101, 102, 103]. Stability of liposome

us
inside the biological fluids is mainly affected by several blood components including lipoproteins
an
(high-density lipoproteins ) and the complement systems [104, 105], disintegration of liposome

membrane formed from dioleoyl phosphatidylethanolamine/oleic acid (DOPA/OA) and leakage of


M
drug content has been reported through the action of albumin [106]. Moreover, calcium ion induces

aggregation of liposome composed from DOPE/OA [107, 108], some studies reported that
e d

incorporation of sphingomyelin in AVE (Artificial Viral Envelopes) liposomes can decrease lysis
pt

occurrence in serum and increase stability [109]. N-acyl-phosphatidyl ethanolamine


ce

incorporation into liposomes has shown to assist the liposomal biological stability

against plasma components and also increase the circulating time of the vesicles
Ac

[110]. Previously published study reported that fluorinated liposomes more stable than the

conventional one, it also indicated that lipophobic characteristics of the fluorinated intramembrane

core protects the liposomes, probably by minimizing their interactions with the lipophilic serum

components decreasing their adsorption at the surface and hindering their diffusion into the

fluorinated bilayer, furthermore, the fluorinated liposomes stability in serum accrue from the higher
negative dipole potential of fluorinated membranes that affect the binding of negatively charged

lipophilic ions reducing the binding of the serum proteins (negatively charged) that penetrate into

the interfacial region of liposomal bilayer [111], moreover, the fluorinated liposomes showed

longer systemic half-lives in comparison with the conventional DSPC and DSPC/cholesterol

liposomes [112]. The binding of polyethylene glycol on the liposome surface was found to decrease

the liposome clearance and reducing the liposome uptake by the phagocyte system [113, 114].

t
ip
Finally, the biological stability of liposome in blood and other biological fluids still needs further

investigation.

cr
us
5 Applications of liposomes

5.1 Liposomes for anticancer drug and gene delivery. an


Due to the nature and behavior of cancer tissue, and the large difference between it and the
M
normal one, the cancer tissue considered an appropriate target for drug delivery system, for

example, the tumor vasculature is characterized by leaky vasculature and limited lymphatic
d

drainage [115], consequently, macromolecules and micromolecules can be easily accumulated in


e
pt

the intercellular spaces of a large variety of tumors. Furthermore, there are specific markers that are

not present in the normal tissue blood vessels (aminopeptidase and N, integrins) [116, 117]. A
ce

continuous angiogenesis in the cancer tissue acts as a target for anticancer drug vectors; once the
Ac

anticancer drug delivery systems reach the extracellular spaces, the active drug can be released by

different mechanisms, such as the pH difference between the blood (7.4) and the extracellular

spaces of cancer cells (4-5 acidic PH), which can affect the acid sensitive polymers carrying the

anticancer drug. Another mechanism is the exposure of anticancer drugs carrying polymers to

degradation by the lysosomal enzyme [118].


Liposomes are used as a carrier for numerous anticancer drugs in the conventional or modified

forms (PEGylated liposome). In a recent study, folic acid (FA)-conjugated liposome was designed

to co-encapsulate both celastrol (Cs) and irinotecan (Ir) and its effect in breast cancer therapy was

investigated. DPPC and DSPE-PEG-NH2 were used for liposome preparation, the formulation

showed small vesicle size (190 nm) with narrow PDI (0.1) and better drug release profile at pH 5.0

for both drugs, furthermore, it exhibited higher cellular uptake and improved apoptosis in breast

t
ip
cancer cells (MCF-7 and MDA-MB-231) [119]. The pharmacokinetic property of doxorubicin was

significantly enhanced and its anticancer effect against human lung carcinoma A549 cells was

cr
improved upon its encapsulation into selenium coated liposome (Dox-SeLPs), lecithin S100,

us
DOTAP, and cholesterol were used for liposomal preparation by ammonium sulfate ((NH4)2SO4)
an
gradient method, Dox-SeLPs displayed higher in-vitro cytotoxicity on A549 cells with significantly

lower IC50 ( 0.92 ± 0.16 μg/mL) than that of free (4.40 ± 0.58 μg/ mL) and liposomal doxorubicin
M
(5.68 ± 0.73 μg/mL) [120]. Antibody fragment (AF)-combined liposome was used for

encapsulation of gemcitabine (GEM) and paclitaxel (PTX). phospholipids used for the preparation
e d

of liposome were DPPC, DSPE-PEG2000, and cholesterol, the drug-loaded liposome displayed a
pt

particle size about 135 nm with a narrow PDI. The cytotoxic effect of the liposomal formulation
ce

(AF-GPL) in pancreatic cancer cells was evaluated, it showed higher cytotoxicity effect with IC50

value of 0.45 μg/ml significantly lower than that of gemcitabine (5.9 μg/ml), paclitaxel (4.2 μg/ml),
Ac

and GEM/ PTX co-loaded liposome (1.92 μg/ml) [121]. cationic liposomes were used as a drug

carrier for STAT3 siRNA and curcumin for topical treatment of skin cancer [122]. Thin film

hydration technique was used for the preparation of liposomes, in which curcumin was mixed with

1,2-dioleoyl-sn-glycero-3-phospho-ethanolamine (DOPE), 1,2-Dioleoyl-3-trimethylammonium

propane (DOTAP), and C6 ceramide. After that, methanol was used to dissolve the mixture. A
rotary evaporator was used to evaporate the solvent, and formation of lipid thin film with the drug,

then using 20 mM buffer at pH 7.4 for film hydration. Finally, complexation of the liposome with

STAT3 siRNA was performed through the addition of curcumin-loaded liposomes to siRNA in the

buffer solution (pH 7.4), then the preparation was vortexed for 30 s, and incubated for 20 min at

room temperature. This preparation was used for growth inhibition of B16F10 Melanoma cells. The

data showed a great inhibition of B16F10 cancer cell growth (76.3±4.0%) by using curcumin-

t
loaded cationic liposome- STAT3 siRNA complex (250 μM curcumin and 0.5 nM siRNA)

ip
compared with individual agents (free STAT3 siRNA and free curcumin). Another study reported

cr
the application of liposomes for improving the penetration of siRNA through the stratum corneum

us
and epidermis for melanoma cells treatment by using the solvent dispersion technique for liposomes
an
preparation [123]. DOTAP solution (1,2-dioleoyl-3-trimethylammonium propane chloride

dissolved in chloroform10 mg/ml) was mixed with a NaChol solution (sodium cholate in ethanol 10
M
mg/ml). Then, the lipid film was formed after the evaporation of the solvent under vacuum,

followed by hydration of the film in buffer (pH 7.4). The data of this study indicated that the
e d

developed lipoplexes had the ability to penetrate the layers of the skin entering basal epidermis cells
pt

and inhibit the target proteins expression. Different lipid material (DOPC/SM/Chol/DOPS/DOPE)
ce

were used for exosome preparation intended for VEGF (vascular endothelial growth factor) siRNA

delivery to human lung carcinoma A549 cells, exosomes exhibited enhanced storage stability and
Ac

anti-serum aggregation effect and also showed significantly higher cellular uptake and silencing

efficacy in comparison with free siRNA and PC-Chol liposomes [124]. The surface modification of

the liposomes such as cationic liposomes was found to be effective Nanocarriers in-vitro, but

unfortunately, the in-vivo study showed cytotoxicity. Therefore, modification of neutral PEGylated

liposomes with cell penetrating peptides (CPPs) was performed to improve the cellular uptake
[125]. In this study, ethanol injection technique was used for liposome preparation, showing

spherical morphology and uni lamellarity of each liposome class with small size vesicles 50-150

nm. The obtained data proved that R8-PLPs (Octaarginine (R8) peptide PEGylated liposomes)

enhanced cellular association, negligible cytotoxicity, and had higher capacity in gene silencing as

compared to the cationic liposomes that showed significant cytotoxicity. Various liposome

formulations for gene delivery and diverse types of anticancer drugs are shown in Table 2.

t
ip
5.2 Applications of liposomes in vaccine formulation

Liposomes are flexible drug delivery systems that can be surface-modified with various

cr
molecules. The encapsulation of peptide antigens or viral membrane proteins into the liposomes

us
has been shown to produce humoral and cell-mediated immune response, and generate solid and
an
durable immunity against the pathogen [126, 127]. For example, Epaxal® vaccine for hepatitis A,

which considered a successful commercial product based on the liposomal formulation as a drug
M
carrier was approved for human use in Switzerland in 1994 [128]. A Recent study explained how

the composition and structure of liposomes demonstrate the impact of the MPER peptides
e d

(membrane-proximal external region A promising HIV antigen) on the strength and durability of
pt

the humoral responses and T-helper cell responses in mice [129]. The MPER of HIV-1 gp41 is a
ce

peptide expressed on the virion surface. It is considered as the most important vaccine recognized

and neutralized by many broadly neutralizing antibody [130]. But this antigen has poor
Ac

immunogenicity and need to be presented on membranes to physiologically matching the HIV

native virus and to enhance the immune response. Liposomes were formulated by mixing [

DOPC(1,2-Dioleoyl-sn-glycero-3-phosphocholine): DOPG (1,2-dioleoyl-sn-glycero-3-phospho-(1′-

rac-glycero1) sodium salt)], palm-MPER and DSPE-PEG in chloroform, then the organic solvent

was evaporated under vacuum, followed by the hydration of the lipid film by using phosphate
buffer. The obtained liposomes undergo vortexing then freeze-thawing cycles, followed by

polycarbonate membranes extrusion. The antigen immunogenicity was affected by various factors,

for instance, immunoglobulin G (IgG) titers specific to MPER is decreased by 15-fold and 20-fold

upon reduction of liposomes particle size from 200 nm and 150 nm, respectively to the particle size

of 65 nm. Also, the immunogenicity is oppositely proportional to the liposome membrane fluidity,

and this confirmed through mice immunization with liposomes with lower membrane fluidity

t
ip
(DMPC (Dimyristoylphosphatidylcoline), DOPC and DOPG) and highly fluid membrane (DOPC

and DOPG) demonstrated that a higher IgG titer about 6.2-fold obtained through immunization by

cr
the DMPC-containing liposome. Furthermore, it was observed that the immunization was enhanced

us
by increasing the density of the antigen on the surface of the liposome while increasing the antigen
an
density to 2000 MPER / liposome led to fewer antibody titers [131].

Another study proved that DPC (dimethyl dioctadecyl ammonium, Poly I: C and
M
cholesterol) can be used as adjuvant forming a unique type of cationic liposome for delivery of
d

Tuberculosis specific antigen protein. The study revealed that the formulation stability was affected
e

upon incorporation of the oppositely charged Poly I: C into liposomes dioctadecyl ammonium
pt

(DDA), and consequently limitation of the clinical application. So, for stability improvement,
ce

cholesterol and gelatin incorporation has been investigated. It was observed that the in-vitro

stability increased along with the addition of gelatin and cholesterol, but unfortunately, the addition
Ac

of gelatin to the AMM TB (subunit vaccine Ag85B-MPT64 (190-198)-Mtb8.4) antigen in adjuvant

to DP resulted in decreasing the protective efficacy of the AMM vaccine. In contrast, cholesterol

addition into DP did not decrease the protective efficacy or the immunogenicity of the vaccine.

Furthermore, the cholesterol addition into the membrane of the liposomes improved the packing of

the lipid, and consequently decreased the phase transition temperature, resulting in in-vivo and in-
vitro stability improvement of the liposomal formulation. Finally, the novel stable adjuvant DPC

had the ability to sensitize the immune response of Th1-type cell [132]. Table 3 shows a liposomal

formulation of different substances used for the generation and potentiation of the immune

response.

5.3 Applications of liposomes in ophthalmology

Drugs applied topically in the form of ointment, solutions, and suspensions are used for the

t
ip
management of most ophthalmic disorders. As a result of various pathophysiological and

cr
anatomical barriers found in the eye, these dosage forms have poor ocular bioavailability. For the

us
management of eye syndromes including the posterior and anterior segments, liposomes were used.

For example, dry eyes have been treated through liposomal aqueous suspension [133], also novel
an
spray has been prepared in which phospholipid liposomes are delivered to the tear film [134].

Liposomal formulation for keratitis has been developed, for example, eye drops of amphotericin B
M
(AmBisome®) would be proper and convenient for fungal infections
d

with Curvularia, Fusarium, Candida, and Aspergillus that can cause corneal serious ulceration
e

[135]. Another formulation of liposomes for corneal transplant rejection, endophthalmitis, uveitis,
pt

and proliferative vitreoretinopathy has been investigated.


ce

Ciprofloxacin (Ciprocin) is a commercially available eye drops, active against both aerobic

gram-positive and gram-negative bacteria. By making a comparison between ciprofloxacin eye


Ac

drops and ciprofloxacin liposomal formulation using the Albino rabbit as an animal model, it was

observed that the maximum aqueous humor concentration (Cmax) was 3.87 mcg/mL from the

liposomal formulation compared with 2.68 mcg/mL for ciprocin eye drops nearly with the same

Tmax. Furthermore, the values of liposome formulations (area under the curve) were significantly

higher than that of ciprocin eye drops, and this refers to the higher ocular absorption, and
consequently bioavailability for ciprofloxacin delivered by liposome in comparison to the eye

drops. As a result, the encapsulation of ciprofloxacin into liposomal formulation leads to higher

bioavailability, residence time enhancement, and reduction in the dose of the drug with a higher

chance for penetration through the tissue of the eye [136]. A novel study made an intercalation

between betaxolol hydrochloride (BH) (an effective drug for glaucoma) and Montmorillonite (Mt)

(ion-exchange drug carrier with the large surface area and perfect exchangeability) and then

t
ip
incorporated into liposomes (Mt-BH-LPs) as an ocular drug-delivery system. A slower release was

observed from Mt-BH-LPs (50.3%) compared with the betaxolol hydrochloride sample that showed

cr
approximately 100% rapid release within 2.5 h. Also, by using the Draize method for the evaluation

us
of the irritation effects of the formulations on the rabbit eye, no irritation effects attributed to Mt-
an
BH-LPs was observed. Furthermore, the in-vitro pre-corneal retention test demonstrated that the

highest BH concentration (38.87μg/mL) was immediately observed and rapidly decreased to


M
undetectable values within 60 min after application of BH sample, while the concentration of

betaxolol reached undetectable values at more than 110 min after the application of Mt-BH-LPs.
e d

Also, the in-vivo test confirmed these in-vitro results, the tears BH concentrations from Mt-BH-LPs
pt

were three-fold larger than those of BH solution within 30 min. Moreover, the concentration of
ce

betaxolol (13.6μg/mL) in Mt-BH-LPs was detected at 240 min. These results together revealed that

Mt-BH-LPs could be able to prolong the drug retention time on the eye surface, and also decreased
Ac

drug elimination [137].

5.4 Liposomes in wound healing

Liposomes were proved to be helpful in enhancing the stability, local deposition and

permeation into the skin and deeper tissues and extending the release of their contents. Furthermore,
it can offer a moist environment on the surface of wound skin because of liposome effective closure

as epidermic cells, which lead to wound healing [138].

There are many substances that cannot penetrate through the skin as a result of their

physicochemical properties, such as hydrophilicity and large molecular weight, which could lead to

lower bioavailability and lower biological response. Using liposomes as drug delivery for these

substances would overcome these problems. External application of growth factors [epidermal

t
ip
growth factor (EGF), fibroblast growth factor (FGFs), vascular endothelial growth factor (VEGF),

and platelet-derived growth factor (PDGF)] has good impacts on wound healing. One of these

cr
growth factors is the basic fibroblast growth factor (bFGF) has the ability to stimulate

us
differentiation and proliferation of fibroblast, and also promote angiogenesis formation, which
an
consequently improves wound healing [139]. Lower stability, short half-life (bFGF about 1.5 min),

and easy degradability (enzymatic degradation) of these growth factors could cause insufficient
M
efficacy. Liposomes have been used to encapsulate bFGF to improve its stability; however, it was

exhibited to rapid leakage when it was topically applied due to the liquid status of liposomes. So,
e d

preparation of liposome with the hydrogel core of silk fibroin including the bFGF growth factor has
pt

been developed to prevent the rapid leakage of the bFGF. By this formulation, bFGF stability in the
ce

wound exudate was maintained, penetration ability was enhanced, and consequently, its cell

proliferation activity and wound healing ability were promoted [140].


Ac

Cysteine protease is an enzyme derived from Carica papaya fruit with higher proteolytic

activity; it promotes the skin exfoliation and produces soft skin through fibrosis removing. But its

large molecular weight and hydrophilic nature limit its use for the topical application. Liposomes

were used for the encapsulation of papain enzyme in order to overcome these drawbacks. By

making a comparison between different papain formulations (papain solution, papain- propylene
glycol physical mixture, and conventional liposomes) and propylene glycol liposomal formulation,

the results approved that the propylene glycol liposomes possessed the higher ability for the

reduction of fibrosis (5.566, 3.509, 2.242 times, respectively). This indicated that the propylene

glycol liposomes increased the deep permeation ability of the enzyme into the skin layers of second

degree burned rats [141]. Various substances encapsulated into liposomes for their application in

wound healing are shown in Table 4.

t
ip
5.5 Application of liposomes in miscellaneous dermatological disorders

cr
Acne vulgaris is a prevalent dermatologic disease with substantial cutaneous and

us
psychological disease burden; it is mostly dispersed in adolescents. The pathogenesis of acne

mainly depends on four factors, stimulation


an of pilosebaceous gland by androgen

(dihydrotestosterone), hyperkeratinization of the follicle, the skin microbial flora colonization


M
(Propionibacterium acnes), and inflammation [142].

Due to the severe systemic side effects of the tretinoin (TRT) [143], it is mostly applied as a
d

topical formulation. However, the effectiveness of TRT as a topical preparation (creams, lotions,
e
pt

gels, and emulsion) is limited because it had higher sensitivity to light and air, and consequently

limited stability and loss during storage. Also, the barrier properties of the skin hinder the
ce

penetration and the deposition of the drug into the skin layers. Accordingly, several studies have
Ac

been carried out to incorporate TRT into liposomes to overcome its problems. Liposomes were

prepared by an ether injection technique since dichloromethane was used to dissolve the lipid

components and TRT, then the solution was injected slowly into distilled water (100 mL) under

stirring condition. Liposomes were also prepared by thin film hydration method, the lipid

component, the drug and cholesterol were dissolved in dichloromethane, and then rotary evaporator

was used to evaporate the solvent followed by hydration the lipid film with distilled water. The
study reported that insignificant erythema (0.2±0.37) was observed for liposomal gel formulation.

In contrast, TRT gel (0.025%) showed significant erythema (1.70±0.751). The marketed product

caused an erythema score of 1.40±0.534 and was not able to reduce the irritation resulted from the

topical application of TRT. The liposomal formulation possessed a zeta potential of - 41.2±1.2mV,

this large value refers to the highest stability of the prepared liposomes since the aggregation of

liposomes was prevented by the strong electrostatic repulsion interaction [144].

t
ip
P. acnes produce extracellular lipase enzyme, a novel study prepared liposome loaded with

cr
lipase-sensitive singlet oxygen-producible (pullulan-pheophorbide a (PU-Pheo A)) and

us
erythromycin (LSSPL). Firstly, the erythromycin liposomes were prepared by the thin film

hydration technique, and then the pullulan-pheophorbide A conjugates (1 mg/mL of Pheo A) was
an
added to the erythromycin liposomes and stirred overnight. The study showed that the lipase

enzyme produced by the P. acnes induced structural breakdown of LSSPL and liberate of the
M
encapsulated erythromycin. It also reported that the using of irradiation by laser onto free Pheo
d

improved the LSSPLs antibacterial activity. The in-vitro study showed that LSSPLs had greater anti
e

bactericidal properties and reduced viability [3.12 log of CFU (99.92%)] compared to OELLs (only
pt

liposome containing erythromycin) [1.99 log of CFU (98.98%)] and OPCLs (liposome coated with
ce

Pheo A) [1.58 log of CFU (97.40%)] under laser irradiation conditions. Reduction in inflammation

caused by the P. acnes in mice skin showed that the OELL application leads to an average skin
Ac

volume decrease to 31.4%, OPCLs treatment using laser irradiation reduced the volume of the skin

by 37.3%, but the LSSPL application with laser irradiation showed superior reduction in the

inflamed skin to 5.5% after 7 days [145].

Psoralen combined with ultraviolet A radiation (PUVA) was recommended by FDA for the

treatment of severe psoriasis. But the lower permeability and poor deposition of psoralen into the
skin layers, and the side effects related to conventional psoralen dosage forms (severe burning,

pigmentation, and blisters) decreased the therapeutic efficiency and safety of topical PUVA.

Anionic and cationic liposome-encapsulated psoralen by thin film hydration technique was

prepared. The ex-vivo test confirmed that cumulative skin permeation of psoralen was improved

about 5-fold difference with liposomal formulation gel [cationic liposome (9.18±0.07%], anionic

liposome [9.28±0.65%)] in comparison to the solution form of psoralen (1.90±0.18%) in 24 h. An

t
ip
in-vivo study in a murine model with psoriasis induced by Imiquimod showed that the mouse

groups treated with liposomal formulation gel showed the highest improvement of psoriasis

cr
symptoms (thickening, erythema, and scales) in comparison to PSR-Solution due to greater skin

us
permeation of psoralen from liposomal carriers [146].
an
Another study made co-encapsulation of resveratrol and psoralen for evaluation of anti-

oxidant and PUVA combination in the management of vitiligo. In this study, mouse melanoma cells
M
(B16F10) was used to evaluate the effect of the prepared formulation due to the low incidence of

vitiligo disease that may cause time-consuming, and the high cost of vitiligo model preparation. The
e d

study observed that co-encapsulation of the two drugs into the ultra-deformable liposomes resulted
pt

in higher stimulation of melanin and tyrosinase activity by psoralen and significant anti-oxidant
ce

activity by resveratrol [147].

6 Present prospective and future challenges


Ac

In the pharmaceutical application, using of liposomes as a drug delivery system for specific

targeting, sustained, and/or controlled release, as well as for immunization, was and still is the

driving force for innovative technologies development. Surface modification of liposomes provides

many targeting approaches and ligand density optimization on liposomes surface can increase the

liposomal uptake in tumor cells. However, increasing the density and the length of the ligand
beyond optimum conditions will lead to aggregation and steric hindrance that will influence ligand-

receptor affinity and interaction. So, the optimum density and length of the ligands and their impact

on the liposomal size, the circulation of liposomes in the blood and the affinity of the ligand to the

targeted receptors should be carefully addressed [148, 149, 150]. Stability of the liposomal

formulation and the leakage of the active components from vesicles after administration are vital

problems which need to be monitored to get the optimum therapeutic effect, especially in tumor

t
ip
therapy. phospholipids used in liposome preparation are very susceptible to oxidation and

hydrolysis, so it has a short shelf life. And also, the electrostatic stabilization of the vesicles cannot

cr
offer acceptable stability to liposomes used for encapsulation of substances highly susceptible to

us
disintegration such as enzymes and proteins intended for in-vivo applications. The liposome
an
solutions sterility is another great challenge that should be considered, sterilization is typically

carried out using filter membranes to avoid degradation caused by other techniques, for instance, γ-
M
irradiation, ultraviolet (UV) and dry heat sterilization, unfortunately, filtration techniques

characterized by more time consuming and lower efficiency for removal of viruses [151], moreover,
e d

because of the higher viscosity of the liposomal preparations that can lead to premature membrane
pt

blocking and also, the lower surface tension of these solutions affects the contact angle with filter
ce

membrane and decreases bubble point increasing the probability of bacterial penetration through

these membranes [152]. Several preparation techniques were developed, however, most of them are
Ac

suitable for small-scale and laboratory application and less for the large-scale production.

Unfortunately, the availability of certain production techniques as well as the quality issues depends

on the properties of the lipids themselves. This limits the choice of liposome kinds from which one

can select when improving liposome-based drug therapy. Severe control of the product quality is

essential to achieve the predictable therapeutic effect, quality control related to unwanted by-
products, such as organic solvents residues, degradation products, lysolipids, and other lipid

detergents are just as vital as pyrogen-free and sterile conditions.

Development of liposome as a drug delivery system to control drug release rates and

enhance disease-specific targeting has shown potential impact in global healthcare. However, there

has been a general deficiency of precise protocols to characterize the liposomal drug product at

physicochemical, physiological and biological levels. The following aspects should be considered

t
ip
to support the development of the liposomal product;

cr
- physicochemical properties of a liposomal product are very important to confirm its quality

us
that usually affected by the purity and stability characteristics of phospholipid and other

critical excipients, the active substance/lipid moiety ratio at appropriate manufacturing steps
an
should be within acceptable range to ensure reliable preparation performance, physical
M
morphology, mean vesicle size and polydispersity index, stability of the active ingredient,

lipid materials and other excipients in the final product, including estimation of degradation
d

products (e.g. lysolipid, hydrolytic/oxidated moieties), in-vitro release rate of the active
e
pt

substance from the vesicles in physiologically/clinically related media [153]. also,

identification and control of main intermediates in the manufacturing process have a critical
ce

impact on the product quality.


Ac

- the non-clinical studies to be carried out prior to clinical studies should include reasonable

study of pharmacokinetics (biodistribution, metabolism, and clearance), pharmacodynamics

and toxicology, during the preclinical evaluation, biocompatibility and immunotoxicity

should be taken into consideration along with an appropriate assessment of product

properties during the development includes administration route, dosage regimen, targeted

disease environment, and therapeutic index.


- development and validation of sensitive analytical methods to quantify drug, metabolite and

other byproducts in blood/tissue will be necessary. The aforementioned criteria along with

the regulatory perspectives of nanomedicine for product development that presented

underlying numerous procedures by the European Medicines Agency (EMA) and Food and

Drug Administration (FDA) can be taken in consideration to support the development and

the marketing approval of liposomal product [154, 155].

t
ip
7 Conclusions

Great attention has been given to liposomes as drug carriers to improve and enhance the

cr
therapeutic index of drugs. Liposomes research has expanded during the last three decades and due

us
to their unique characteristics (non-toxic, biodegradable, biocompatible, non-immunogenic, lowers
an
systemic toxicity, sensitive drug molecules protection, targeted delivery, and improved

pharmacokinetic properties). Liposomes have a wide range of functions and applications including
M
the using of more precise cell ligand-targeted on their surface due to the facile change of structure.

The main challenge is that it is impossible to cross most regular pellicle barriers due to their
e d

imposed size. However, with the advance of liposomes technology, they will play a more important
pt

functional role in the clinical environment. Improving the future researches on the existing
ce

platforms and to address the current translational and regulatory limitations would be achieved by

understanding the advances and developments in liposomes technology, and the overcoming their
Ac

formulation challenges.

Conflict of interest

The authors declare no conflict of interest.


Acknowledgments

This work was supported by the National Natural Science Foundation of China (81503007

and 21574059).

References

t
1. Petersen GH, Alzghari SK, Chee W, et al. Meta-analysis of clinical and preclinical studies comparing

ip
the anticancer efficacy of liposomal versus conventional non-liposomal doxorubicin. Journal of
Controlled Release. 2016;232:255-264.

cr
2. Ito K, Hamamichi S, Asano M, et al. Radiolabeled liposome imaging determines an indication for
liposomal anticancer agent in ovarian cancer mouse xenograft models. Cancer science.
2016;107(1):60-67.

us
3. Drulis-Kawa Z, Dorotkiewicz-Jach A. Liposomes as delivery systems for antibiotics. International
Journal of Pharmaceutics. 2010 2010/03/15/;387(1):187-198. doi:
https://doi.org/10.1016/j.ijpharm.2009.11.033. an
4. Jung SW, Thamphiwatana S, Zhang L, et al. Mechanism of Antibacterial Activity of Liposomal
Linolenic Acid against Helicobacter pylori. PloS one. 2015;10(3):e0116519. doi:
10.1371/journal.pone.0116519.
M
5. Ghanbarzadeh S, Arami S. Enhanced Transdermal Delivery of Diclofenac Sodium via Conventional
Liposomes, Ethosomes, and Transfersomes. BioMed Research International. 2013 07/14
04/29/received 06/16/revised 06/17/accepted;2013:616810. doi: 10.1155/2013/616810. PubMed
d

PMID: PMC3725948.
e

6. Fujisawa T, Miyai H, Hironaka K, et al. Liposomal diclofenac eye drop formulations targeting the
retina: Formulation stability improvement using surface modification of liposomes. International
pt

Journal of Pharmaceutics. 2012 2012/10/15/;436(1):564-567. doi:


https://doi.org/10.1016/j.ijpharm.2012.07.024.
ce

7. Teagle AR, Birchall JC, Hargest R. Gene therapy for pyoderma gangrenosum: optimal transfection
conditions and effect of drugs on gene delivery in the HaCaT cell line using cationic liposomes. Skin
pharmacology and physiology. 2016;29(3):119-129.
8. Zylberberg C, Gaskill K, Pasley S, et al. Engineering liposomal nanoparticles for targeted gene
Ac

therapy. Gene Therapy. 2017.


9. Perez AP, Altube MJ, Schilrreff P, et al. Topical amphotericin B in ultradeformable liposomes:
formulation, skin penetration study, antifungal and antileishmanial activity in vitro. Colloids and
Surfaces B: Biointerfaces. 2016;139:190-198.
10. Della Pepa R, Picardi M, Sorà F, et al. Successful management of chronic disseminated candidiasis in
hematologic patients treated with high-dose liposomal amphotericin B: a retrospective study of the
SEIFEM registry. Supportive Care in Cancer. 2016;24(9):3839-3845.
11. Allen TM, Moase EH. Therapeutic opportunities for targeted liposomal drug delivery. Advanced
Drug Delivery Reviews. 1996 1996/09/16/;21(2):117-133. doi: https://doi.org/10.1016/S0169-
409X(96)00402-4.
12. Ming-Kung Y. Clinically-proven liposome-based drug delivery: formulation, characterization and
therapeutic efficacy. Open Access Scientific Reports. 2012.
13. Makino K, Shibata A. Surface properties of liposomes depending on their composition. Advances in
Planar Lipid Bilayers and Liposomes. 2006;4:49-77.
14. Jesorka A, Orwar O. Liposomes: technologies and analytical applications. Annu Rev Anal Chem.
2008;1:801-832.
15. Akbarzadeh A, Rezaei-Sadabady R, Davaran S, et al. Liposome: classification, preparation, and
applications [journal article]. Nanoscale Research Letters. 2013 February 22;8(1):102. doi:
10.1186/1556-276x-8-102.
16. Shehata T, Ogawara K-i, Higaki K, et al. Prolongation of residence time of liposome by surface-
modification with mixture of hydrophilic polymers. International Journal of Pharmaceutics. 2008
2008/07/09/;359(1):272-279. doi: https://doi.org/10.1016/j.ijpharm.2008.04.004.

t
17. Biswas S, Dodwadkar NS, Sawant RR, et al. Surface modification of liposomes with rhodamine-123-

ip
conjugated polymer results in enhanced mitochondrial targeting. Journal of drug targeting. 2011
02/25;19(7):552-561. doi: 10.3109/1061186X.2010.536983. PubMed PMID: PMC3492939.

cr
18. Zhou J, Zhao W-Y, Ma X, et al. The anticancer efficacy of paclitaxel liposomes modified with
mitochondrial targeting conjugate in resistant lung cancer. Biomaterials. 2013;34(14):3626-3638.
19. Paltauf F, Hermetter A. Phospholipids — Natural, Semisynthetic, Synthetic. In: Hanin I, Pepeu G,

us
editors. Phospholipids: Biochemical, Pharmaceutical, and Analytical Considerations. Boston, MA:
Springer US; 1990. p. 1-12.
20. Li J, Wang X, Zhang T, et al. A review on phospholipids and their main applications in drug delivery
an
systems. Asian Journal of Pharmaceutical Sciences. 2015 2015/04/01/;10(2):81-98. doi:
https://doi.org/10.1016/j.ajps.2014.09.004.
21. D'Avanzo N. Chapter Twelve - Lipid Regulation of Sodium Channels. In: French RJ, Noskov SY,
M
editors. Current Topics in Membranes. Vol. 78: Academic Press; 2016. p. 353-407.
22. McIntosh TJ, Simon SA, Needham D, et al. Structure and cohesive properties of
sphingomyelin/cholesterol bilayers. Biochemistry. 1992;31(7):2012-2020.
d

23. Carter KA, Luo D, Razi A, et al. Sphingomyelin Liposomes Containing Porphyrin-phospholipid for
Irinotecan Chemophototherapy. Theranostics. 2016 10/01 03/30/received
e

06/24/accepted;6(13):2329-2336. doi: 10.7150/thno.15701. PubMed PMID: PMC5118598.


pt

24. Chiho Uemori TK, Siti Machmudah, Wahyudiono, Hideki, Kanda KY, Motonobu Goto. Production of
Liposome from Sphingomyelin by Ultrasonic Device under Supercritical Carbon Dioxide. Asian
ce

Journal of Applied Sciences,. 2017;5:1042-1048.

25. Sim JS, Nakai S. Egg uses and processing technologies: new developments. Cab International; 1994.
26. Wang H, Yao L, Lee S-L, et al. Extraction of Phospholipids from Egg Yolk Flakes Using Aqueous
Ac

Alcohols. Journal of the American Oil Chemists' Society. 2017;94(2):309-314.


27. Wang H, Yao L, Lee SL, et al. Simultaneous texturization and extraction of phospholipids from liquid
egg yolk using renewable solvents. European Journal of Lipid Science and Technology. 2017;119(2).
28. Liu Z-Y, Zhou D-Y, Wu Z-X, et al. Extraction and detailed characterization of phospholipid-enriched
oils from six species of edible clams. Food Chemistry. 2018;239:1175-1181.
29. Ali AH, Zou X, Lu J, et al. Identification of phospholipids classes and molecular species in different
types of egg yolk by using UPLC-Q-TOF-MS. Food chemistry. 2017;221:58-66.
30. Walczak J, Bocian S, Kowalkowski T, et al. Determination of Omega Fatty Acid Profiles in Egg Yolk by
HILIC-LC-MS and GC-MS. Food Analytical Methods. 2017;10(5):1264-1272.
31. Belayneh HD, Wehling RL, Reddy AK, et al. Ethanol-Modified Supercritical Carbon Dioxide
Extraction of the Bioactive Lipid Components of Camelina sativa Seed. Journal of the American Oil
Chemists' Society. 2017;94(6):855-865.
32. Samad A, Sultana Y, Aqil M. Liposomal drug delivery systems: an update review. Current drug
delivery. 2007;4(4):297-305.
33. Bangham A, Standish MM, Watkins J. Diffusion of univalent ions across the lamellae of swollen
phospholipids. Journal of molecular biology. 1965;13(1):238-IN27.
34. Bhatia A, Kumar R, Katare OP. Tamoxifen in topical liposomes: development, characterization and
in-vitro evaluation. J Pharm Pharm Sci. 2004;7(2):252-259.
35. Ghanbarzadeh S, Valizadeh H, Zakeri-Milani P. Application of response surface methodology in
development of sirolimus liposomes prepared by thin film hydration technique. BioImpacts: BI.
2013;3(2):75.

t
36. Mathai JC, Sitaraman V. Preparation of large uni-lamellar liposomes by the ether injection method

ip
and evaluation of the physical integrity by osmometry. Biochemical Education. 1987;15(3):147-149.
37. Charcosset C, Juban A, Valour J-P, et al. Preparation of liposomes at large scale using the ethanol

cr
injection method: Effect of scale-up and injection devices. Chemical Engineering Research and
Design. 2015;94:508-515.
38. Jaafar-Maalej C, Diab R, Andrieu V, et al. Ethanol injection method for hydrophilic and lipophilic

us
drug-loaded liposome preparation. Journal of liposome research. 2010;20(3):228-243.
39. Sriwongsitanont S, Ueno M. Effect of freeze-thawing process on the size and lamellarity of peg-lipid
liposomes. The Open Colloid Science Journal. 2010;4(1).
an
40. Costa AP, Xu X, Burgess DJ. Freeze-anneal-thaw cycling of unilamellar liposomes: effect on
encapsulation efficiency. Pharmaceutical research. 2014;31(1):97-103.
41. Mayer L, Hope M, Cullis P, et al. Solute distributions and trapping efficiencies observed in freeze-
M
thawed multilamellar vesicles. Biochimica et Biophysica Acta (BBA)-Biomembranes.
1985;817(1):193-196.
42. Mugabe C, Azghani AO, Omri A. Preparation and characterization of dehydration–rehydration
d

vesicles loaded with aminoglycoside and macrolide antibiotics. International Journal of


Pharmaceutics. 2006 2006/01/13/;307(2):244-250. doi:
e

https://doi.org/10.1016/j.ijpharm.2005.10.005.
43. Gregoriadis G, Leathwood PD, Ryman BE. Enzyme entrapment in liposomes. FEBS Letters. 1971
pt

1971/04/01/;14(2):95-99. doi: https://doi.org/10.1016/0014-5793(71)80109-6.


44. Gregoriadis G, da Silva H, Florence AT. A procedure for the efficient entrapment of drugs in
ce

dehydration-rehydration liposomes (DRVs). International Journal of Pharmaceutics. 1990


1990/11/30/;65(3):235-242. doi: https://doi.org/10.1016/0378-5173(90)90148-W.
45. Budisa N, Schulze-Makuch D. Supercritical Carbon Dioxide and Its Potential as a Life-Sustaining
Ac

Solvent in a Planetary Environment. Life. 2014;4(3). doi: 10.3390/life4030331.


46. Lesoin L, Crampon C, Boutin O, et al. Preparation of liposomes using the supercritical anti-solvent
(SAS) process and comparison with a conventional method. The Journal of Supercritical Fluids. 2011
2011/06/01/;57(2):162-174. doi: https://doi.org/10.1016/j.supflu.2011.01.006.
47. Trucillo P, Campardelli R, Reverchon E. Production of liposomes loaded with antioxidants using a
supercritical CO2 assisted process. Powder Technology. 2018 2018/01/01/;323:155-162. doi:
https://doi.org/10.1016/j.powtec.2017.10.007.
48. Otake K, Shimomura T, Goto T, et al. Preparation of liposomes using an improved supercritical
reverse phase evaporation method. Langmuir. 2006 Mar 14;22(6):2543-50. doi:
10.1021/la051654u. PubMed PMID: 16519453; eng.
49. Santo IE, Campardelli R, Albuquerque EC, et al. Liposomes preparation using a supercritical fluid
assisted continuous process. Chemical Engineering Journal. 2014 2014/08/01/;249:153-159. doi:
https://doi.org/10.1016/j.cej.2014.03.099.
50. Misra A, Jinturkar K, Patel D, et al. Recent advances in liposomal dry powder formulations:
preparation and evaluation. Expert Opinion on Drug Delivery. 2009 2009/01/01;6(1):71-89. doi:
10.1517/17425240802652309.
51. Chatterjee S, Banerjee DK. Preparation, Isolation, and Characterization of Liposomes Containing
Natural and Synthetic Lipids. In: Basu SC, Basu M, editors. Liposome Methods and Protocols.
Totowa, NJ: Humana Press; 2002. p. 3-16.
52. Maulucci G, De Spirito M, Arcovito G, et al. Particle Size Distribution in DMPC Vesicles Solutions
Undergoing Different Sonication Times. Biophysical Journal. 2005 02/04 08/03/received
01/10/accepted;88(5):3545-3550. doi: 10.1529/biophysj.104.048876. PubMed PMID:

t
PMC1305501.

ip
53. Patty PJ, Frisken BJ. The Pressure-Dependence of the Size of Extruded Vesicles. Biophysical

cr
Journal.85(2):996-1004. doi: 10.1016/S0006-3495(03)74538-X.
54. Ong SGM, Chitneni M, Lee KS, et al. Evaluation of Extrusion Technique for Nanosizing Liposomes.
Pharmaceutics. 2016 12/2105/28/received 12/06/accepted;8(4):36. doi:

us
10.3390/pharmaceutics8040036. PubMed PMID: PMC5198018.

55. Wagner A, Vorauer-Uhl K. Liposome Technology for Industrial Purposes. Journal of Drug Delivery.

56.
2011;2011. doi: 10.1155/2011/591325.
an
Barnadas-Rodriguez R, Sabes M. Factors involved in the production of liposomes with a high-
pressure homogenizer. Int J Pharm. 2001 Feb 1;213(1-2):175-86. PubMed PMID: 11165105; eng.
57. Baalousha M, Stolpe B, Lead JR. Flow field-flow fractionation for the analysis and characterization
M
of natural colloids and manufactured nanoparticles in environmental systems: a critical review.
Journal of chromatography A. 2011 Jul 8;1218(27):4078-103. doi: 10.1016/j.chroma.2011.04.063.
PubMed PMID: 21621214; eng.
d

58. Borghi F. Flow Field–Flow Fractionation for size analysis and characterization of nanoparticles for
applications in Life Sciences: alma; 2014.
e

59. Kaszuba M, McKnight D, Connah MT, et al. Measuring sub nanometre sizes using dynamic light
pt

scattering. Journal of Nanoparticle Research. 2008 2008/05/01;10(5):823-829. doi:


10.1007/s11051-007-9317-4.
60. Mohr K, Müller SS, Müller LK, et al. Evaluation of multifunctional liposomes in human blood serum
ce

by light scattering. Langmuir. 2014;30(49):14954-14962.


61. Bhattacharjee S. DLS and zeta potential–What they are and what they are not? Journal of
Controlled Release. 2016;235:337-351.
Ac

62. Ong SGM, Ming LC, Lee KS, et al. Influence of the encapsulation efficiency and size of liposome on
the oral bioavailability of griseofulvin-loaded liposomes. Pharmaceutics. 2016;8(3):25.
63. Fetih G, Fathalla D, El‐Badry M. Liposomal gels for site‐specific, sustained delivery of celecoxib: In
vitro and in vivo evaluation. Drug development research. 2014;75(4):257-266.
64. Camacho KM, Menegatti S, Vogus DR, et al. DAFODIL: a novel liposome-encapsulated synergistic
combination of doxorubicin and 5FU for low dose chemotherapy. Journal of Controlled Release.
2016;229:154-162.
65. Erdoğ A, Limasale P, Dwi Y, et al. In Vitro Characterization of a Liposomal Formulation of Celecoxib
Containing 1, 2‐Distearoyl‐sn‐Glycero‐3‐Phosphocholine, Cholesterol, and Polyethylene Glycol and
its Functional Effects Against Colorectal Cancer Cell Lines. Journal of pharmaceutical sciences.
2013;102(10):3666-3677.
66. Deniz A, Sade A, Severcan F, et al. Celecoxib-loaded liposomes: effect of cholesterol on
encapsulation and in vitro release characteristics. Bioscience reports. 2010;30(5):365-373.
67. Moghimipour E, Salami A, Monjezi M. Formulation and Evaluation of Liposomes for Transdermal
Delivery of Celecoxib. Jundishapur journal of natural pharmaceutical products. 2015;10(1).
68. Jain SK, Gupta Y, Jain A, et al. Multivesicular liposomes bearing celecoxib-β-cyclodextrin complex
for transdermal delivery. Drug delivery. 2007;14(6):327-335.
69. Moghimipour E, Handali S. Utilization of thin film method for preparation of celecoxib loaded
liposomes. Advanced pharmaceutical bulletin. 2012;2(1):93.
70. Yamamoto E, Miyazaki S, Aoyama C, et al. A simple and rapid measurement method of
encapsulation efficiency of doxorubicin loaded liposomes by direct injection of the liposomal
suspension to liquid chromatography. International journal of pharmaceutics. 2017.
71. Ohnishi N, Yamamoto E, Tomida H, et al. Rapid determination of the encapsulation efficiency of a

t
liposome formulation using column-switching HPLC. International journal of pharmaceutics.

ip
2013;441(1):67-74.
72. Ran C, Chen D, Xu M, et al. A study on characteristic of different sample pretreatment methods to

cr
evaluate the entrapment efficiency of liposomes. Journal of Chromatography B. 2016;1028:56-62.
73. Briuglia M-L, Rotella C, McFarlane A, et al. Influence of cholesterol on liposome stability and on in
vitro drug release. Drug delivery and translational research. 2015;5(3):231-242.

us
74. Plourde K, Derbali RM, Desrosiers A, et al. Aptamer-based liposomes improve specific drug loading
and release. J Control Release. 2017 Apr 10;251:82-91. doi: 10.1016/j.jconrel.2017.02.026. PubMed
PMID: 28238787; eng. an
75. Fritze A, Hens F, Kimpfler A, et al. Remote loading of doxorubicin into liposomes driven by a
transmembrane phosphate gradient. Biochimica et Biophysica Acta (BBA) - Biomembranes. 2006
2006/10/01/;1758(10):1633-1640. doi: https://doi.org/10.1016/j.bbamem.2006.05.028.
M
76. Zucker D, Marcus D, Barenholz Y, et al. Liposome drugs' loading efficiency: A working model based
on loading conditions and drug's physicochemical properties. Journal of Controlled Release. 2009
2009/10/01/;139(1):73-80. doi: https://doi.org/10.1016/j.jconrel.2009.05.036.
d

77. Briuglia ML, Rotella C, McFarlane A, et al. Influence of cholesterol on liposome stability and on in
vitro drug release. Drug delivery and translational research. 2015 Jun;5(3):231-42. doi:
e

10.1007/s13346-015-0220-8. PubMed PMID: 25787731; eng.


78. Sadeghi N, Deckers R, Ozbakir B, et al. Influence of cholesterol inclusion on the doxorubicin release
pt

characteristics of lysolipid-based thermosensitive liposomes. International journal of


pharmaceutics. 2017 Nov 7. doi: 10.1016/j.ijpharm.2017.11.002. PubMed PMID: 29126907; eng.
ce

79. Fang YP, Hu PY, Huang YB. Diminishing the side effect of mitomycin C by using pH-sensitive
liposomes: in vitro characterization and in vivo pharmacokinetics. Drug design, development and
therapy. 2018;12:159-169. doi: 10.2147/dddt.s150201. PubMed PMID: 29391780; PubMed Central
Ac

PMCID: PMCPmc5774480. eng.


80. Okazaki S, Iwasaki T, Yuba E, et al. Evaluation of pH-sensitive fusogenic polymer-modified
liposomes co-loaded with antigen and alpha-galactosylceramide as an anti-tumor vaccine. The
Journal of veterinary medical science. 2017 Dec 28. doi: 10.1292/jvms.17-0491. PubMed PMID:
29311431; eng.
81. Miyazaki M, Yuba E. Hyaluronic Acid-Based pH-Sensitive Polymer-Modified Liposomes for Cell-
Specific Intracellular Drug Delivery Systems. 2018 Jan 17;29(1):44-55. doi:
10.1021/acs.bioconjchem.7b00551. PubMed PMID: 29183110.
82. Yan X, An X. Thermal and photic stimuli-responsive polydiacetylene liposomes with reversible
fluorescence. Nanoscale. 2013 Jul 21;5(14):6280-3. doi: 10.1039/c3nr00954h. PubMed PMID:
23740098; eng.
83. Kim MS, Lee DW, Park K, et al. Temperature-triggered tumor-specific delivery of anticancer agents
by cRGD-conjugated thermosensitive liposomes. Colloids and surfaces B, Biointerfaces. 2014 Apr
1;116:17-25. doi: 10.1016/j.colsurfb.2013.12.045. PubMed PMID: 24441178; eng.
84. Park SM, Kim MS, Park SJ, et al. Novel temperature-triggered liposome with high stability:
formulation, in vitro evaluation, and in vivo study combined with high-intensity focused ultrasound
(HIFU). Journal of controlled release : official journal of the Controlled Release Society. 2013 Sep
28;170(3):373-9. doi: 10.1016/j.jconrel.2013.06.003. PubMed PMID: 23770213; eng.
85. Mathiyazhakan M, Yang Y, Liu Y, et al. Non-invasive controlled release from gold nanoparticle
integrated photo-responsive liposomes through pulse laser induced microbubble cavitation.
Colloids and surfaces B, Biointerfaces. 2015 Feb 1;126:569-74. doi: 10.1016/j.colsurfb.2014.11.019.
PubMed PMID: 25481686; eng.
86. Mathiyazhakan M, Chan W, Ohl CD, et al. Synthesis of Gold Nanoparticle Integrated Photo-

t
responsive Liposomes and Measurement of Their Microbubble Cavitation upon Pulse Laser

ip
Excitation. Journal of visualized experiments : JoVE. 2016 Feb 24(108):53619. doi: 10.3791/53619.
PubMed PMID: 26967978; eng.

cr
87. Hansen AH, Mouritsen OG, Arouri A. Enzymatic action of phospholipase A(2) on liposomal drug
delivery systems. International journal of pharmaceutics. 2015 Aug 1;491(1-2):49-57. doi:
10.1016/j.ijpharm.2015.06.005. PubMed PMID: 26056930; eng.

us
88. Sangtani A, Petryayeva E, Wu M, et al. Intracellularly Actuated Quantum Dot-Peptide-Doxorubicin
Nanobioconjugates for Controlled Drug Delivery via the Endocytic Pathway. 2018 Jan 17;29(1):136-
148. doi: 10.1021/acs.bioconjchem.7b00658. PubMed PMID: 29191007.
an
89. Fan Y, Wang Q, Lin G, et al. Combination of using prodrug-modified cationic liposome
nanocomplexes and a potentiating strategy via targeted co-delivery of gemcitabine and docetaxel
for CD44-overexpressed triple negative breast cancer therapy. Acta biomaterialia. 2017 Oct
M
15;62:257-272. doi: 10.1016/j.actbio.2017.08.034. PubMed PMID: 28859899; eng.
90. Kono Y, Nakai T, Taguchi H, et al. Development of magnetic anionic liposome/atelocollagen
complexes for efficient magnetic drug targeting. Drug delivery. 2017 Nov;24(1):1740-1749. doi:
d

10.1080/10717544.2017.1402219. PubMed PMID: 29141461; eng.


91. Zhu HM, Gu JH, Xie Y, et al. Hydroxycamptothecin liposomes based on thermal and magnetic dual-
e

responsive system: preparation, in vitro and in vivo antitumor activity, microdialysis-based tumor
pharmacokinetics. J Drug Target. 2018 Apr;26(4):345-356. doi: 10.1080/1061186x.2017.1380654.
pt

PubMed PMID: 28920483; eng.


92. Spera R, Apollonio F, Liberti M, et al. Controllable release from high-transition temperature
ce

magnetoliposomes by low-level magnetic stimulation. Colloids and surfaces B, Biointerfaces. 2015


Jul 1;131:136-40. doi: 10.1016/j.colsurfb.2015.04.030. PubMed PMID: 26042528; eng.
93. Patel VR, Agrawal YK. Nanosuspension: An approach to enhance solubility of drugs. Journal of
Ac

Advanced Pharmaceutical Technology & Research. 2011 Apr-Jun;2(2):81-87. doi: 10.4103/2231-


4040.82950. PubMed PMID: PMC3217698.
94. Bhattacharjee S. DLS and zeta potential - What they are and what they are not? Journal of
controlled release : official journal of the Controlled Release Society. 2016 Aug 10;235:337-351.
doi: 10.1016/j.jconrel.2016.06.017. PubMed PMID: 27297779; eng.
95. Staudenecker J. Studies on the Manufacture & Spray Drying of Liposomes and Membrane Lipids
2016.
96. Nounou M, El-Khordagui LK, Khalafallah N, et al. Influence of different sugar cryoprotectants on the
stability and physicochemical characteristics of freeze-dried 5-fluorouracil plurilamellar vesicles.
Vol. 13. 2005.
97. Yang S, Liu C, Liu W, et al. Preparation and Characterization of Nanoliposomes Entrapping Medium-
Chain Fatty Acids and Vitamin C by Lyophilization. International Journal of Molecular Sciences. 2013
09/30 07/15/received 09/12/revised 09/22/accepted;14(10):19763-19773. doi:
10.3390/ijms141019763. PubMed PMID: PMC3821584.

98. Hua Z-Z, Li B-G, Liu Z-J, et al. Freeze-Drying of Liposomes with Cryoprotectants and Its Effect on
Retention Rate of Encapsulated Ftorafur and Vitamin A. Drying Technology. 2003
2003/01/10;21(8):1491-1505. doi: 10.1081/DRT-120024489.
99. Sylvester B, Porfire A, Van Bockstal PJ, et al. Formulation Optimization of Freeze-Dried Long-
Circulating Liposomes and In-Line Monitoring of the Freeze-Drying Process Using an NIR
Spectroscopy Tool. J Pharm Sci. 2018 Jan;107(1):139-148. doi: 10.1016/j.xphs.2017.05.024.
PubMed PMID: 28551424; eng.
100. Rushmi ZT, Akter N, Mow RJ, et al. The impact of formulation attributes and process parameters on
black seed oil loaded liposomes and their performance in animal models of analgesia. Saudi

t
Pharmaceutical Journal. 2017 2017/03/01/;25(3):404-412. doi:

ip
https://doi.org/10.1016/j.jsps.2016.09.011.
101. El-Nesr OH, Yahiya SA, El-Gazayerly ON. Effect of formulation design and freeze-drying on

cr
properties of fluconazole multilamellar liposomes. Saudi Pharmaceutical Journal. 2010
2010/10/01/;18(4):217-224. doi: https://doi.org/10.1016/j.jsps.2010.07.003.
102. Thor Ingvarsson P, Yang M, Nielsen H, et al. Stabilization of liposomes during drying. Vol. 8. 2011.

us
103. Franzé S SF, Samaritani E, Minghetti P, Cilurzo F Lyophilization of Liposomal Formulations: Still
Necessary, Still Challenging. . Pharmaceutics. 2018:10(3):139.
104. Chonn A, Cullis PR, Devine DV. The role of surface charge in the activation of the classical and
an
alternative pathways of complement by liposomes. The Journal of Immunology.
1991;146(12):4234.
105. Tall AR, Tabas I, Williams KJ. [37] Lipoprotein-liposome interactions. Methods in Enzymology. Vol.
M
128: Academic Press; 1986. p. 647-657.
106. Liu D, Huang L, Moore MA, et al. Interactions of serum proteins with small unilamellar liposomes
composed of dioleoylphosphatidylethanolamine and oleic acid: high-density lipoprotein,
apolipoprotein A1, and amphipathic peptides stabilize liposomes. Biochemistry. 1990 Apr
d

17;29(15):3637-43. PubMed PMID: 2111162; eng.


e

107. Fatouros DG, Piperoudi S, Gortzi O, et al. Physical Stability of Sonicated Arsonoliposomes: Effect of
Calcium Ions. Journal of Pharmaceutical Sciences. 2005 2005/01/01/;94(1):46-55. doi:
pt

https://doi.org/10.1002/jps.20221.
108. Collins D, Connor J, Ting-Beall H-P, et al. Proton and divalent cations induce synergistic but
ce

mechanistically different destabilizations of pH-sensitive liposomes composed of dioleoyl


phosphatidylethanolamine and oleic acid. Chemistry and Physics of Lipids. 1990
1990/09/01/;55(3):339-349. doi: https://doi.org/10.1016/0009-3084(90)90172-N.
109. Mady M. SERUM STABILITY OF NON-CATIONIC LIPOSOMES USED FOR DNA DELIVERY. 2018.
Ac

110. Vermehren C, Hansen HS, Clausen-Beck B, et al. In vitro and in vivo aspects of N-acyl-
phosphatidylethanolamine-containing liposomes. International journal of pharmaceutics. 2003 Mar
18;254(1):49-53. PubMed PMID: 12615408; eng.
111. Clary L, Verderone G, Santaella C, et al. Membrane permeability and stability of liposomes made
from highly fluorinated double-chain phosphocholines derived from diaminopropanol, serine or
ethanolamine. Biochimica et Biophysica Acta (BBA) - Biomembranes. 1997
1997/08/14/;1328(1):55-64. doi: https://doi.org/10.1016/S0005-2736(97)00076-X.
112. Santaella C, Frézard F, Vierling P, et al. Extended in vivo blood circulation time of fluorinated
liposomes. FEBS Letters. 1993 1993/12/28/;336(3):481-484. doi: https://doi.org/10.1016/0014-
5793(93)80860-W.
113. Gasselhuber A, Dreher MR, Rattay F, et al. Comparison of Conventional Chemotherapy, Stealth
Liposomes and Temperature-Sensitive Liposomes in a Mathematical Model. PLOS ONE.
2012;7(10):e47453. doi: 10.1371/journal.pone.0047453.
114. Sahil K, Premjeet S, Bilandi A, et al. Stealth liposomes: a review. Vol. 2. 2011.
115. Maeda H, Wu J, Sawa T, et al. Tumor vascular permeability and the EPR effect in macromolecular
therapeutics: a review. J Control Release. 2000 Mar 01;65(1-2):271-84. PubMed PMID: 10699287;
eng.
116. Hallahan D, Geng L, Qu S, et al. Integrin-mediated targeting of drug delivery to irradiated tumor
blood vessels. Cancer Cell. 2003 2003/01//;3(1):63-74. doi: 10.1016/s1535-6108(02)00238-6.
PubMed PMID: 12559176; eng.
117. Pasqualini R, Koivunen E, Kain R, et al. Aminopeptidase N is a receptor for tumor-homing peptides
and a target for inhibiting angiogenesis. Cancer Res. 2000 Feb 01;60(3):722-7. PubMed PMID:

t
10676659; PubMed Central PMCID: PMCPmc4469333. eng.

ip
118. Ulbrich K, Šubr Vr. Polymeric anticancer drugs with pH-controlled activation. Advanced drug
delivery reviews. 2004;56(7):1023-1050.

cr
119. Soe ZC, Thapa RK, Ou W, et al. Folate receptor-mediated celastrol and irinotecan combination
delivery using liposomes for effective chemotherapy. Colloids and Surfaces B: Biointerfaces. 2018
2018/10/01/;170:718-728. doi: https://doi.org/10.1016/j.colsurfb.2018.07.013.

us
120. Xie Q, Deng W, Yuan X, et al. Selenium-functionalized liposomes for systemic delivery of
doxorubicin with enhanced pharmacokinetics and anticancer effect. European Journal of
Pharmaceutics and Biopharmaceutics. 2018 2018/01/01/;122:87-95. doi:
an
https://doi.org/10.1016/j.ejpb.2017.10.010.
121. Yang W, Hu Q, Xu Y, et al. Antibody fragment-conjugated gemcitabine and paclitaxel-based
liposome for effective therapeutic efficacy in pancreatic cancer. Materials Science and Engineering:
M
C. 2018 2018/08/01/;89:328-335. doi: https://doi.org/10.1016/j.msec.2018.04.011.
122. Jose A, Labala S, Ninave KM, et al. Effective Skin Cancer Treatment by Topical Co-delivery of
Curcumin and STAT3 siRNA Using Cationic Liposomes. AAPS PharmSciTech. 2017:1-10.
d

123. Dorrani M, Garbuzenko OB, Minko T, et al. Development of edge-activated liposomes for siRNA
delivery to human basal epidermis for melanoma therapy. Journal of Controlled Release.
e

2016;228:150-158.
124. Lu M, Zhao X, Xing H, et al. Comparison of exosome-mimicking liposomes with conventional
pt

liposomes for intracellular delivery of siRNA. International journal of pharmaceutics. 2018


2018/10/25/;550(1):100-113. doi: https://doi.org/10.1016/j.ijpharm.2018.08.040.
ce

125. Fisher RK, Mattern-Schain SI, Best MD, et al. Improving the efficacy of liposome-mediated vascular
gene therapy via lipid surface modifications. Journal of Surgical Research. 2017;219:136-144.
126. Schwendener RA. Liposomes as vaccine delivery systems: a review of the recent advances.
Ac

Therapeutic advances in vaccines. 2014;2(6):159-182.


127. Zhao L, Seth A, Wibowo N, et al. Nanoparticle vaccines. Vaccine. 2014 2014/01/09/;32(3):327-337.
doi: https://doi.org/10.1016/j.vaccine.2013.11.069.
128. Bovier PA. Epaxal®: a virosomal vaccine to prevent hepatitis A infection. Expert review of vaccines.
2008;7(8):1141-1150.
129. Hanson MC, Abraham W, Crespo MP, et al. Liposomal vaccines incorporating molecular adjuvants
and intrastructural T-cell help promote the immunogenicity of HIV membrane-proximal external
region peptides. Vaccine. 2015 2/11/;33(7):861-868. doi:
https://doi.org/10.1016/j.vaccine.2014.12.045.
130. Ofek G, Zirkle B, Yang Y, et al. Structural basis for HIV-1 neutralization by 2F5-like antibodies m66
and m66.6. Journal of virology. 2014 Mar;88(5):2426-41. doi: 10.1128/jvi.02837-13. PubMed PMID:
24335316; PubMed Central PMCID: PMCPmc3958054. eng.
131. Hanson MC, Abraham W, Crespo MP, et al. Liposomal vaccines incorporating molecular adjuvants
and intrastructural T-cell help promote the immunogenicity of HIV membrane-proximal external
region peptides. Vaccine. 2015 Feb 11;33(7):861-8. doi: 10.1016/j.vaccine.2014.12.045. PubMed
PMID: 25559188; PubMed Central PMCID: PMCPmc4312707. eng.
132. Liu X, Da Z, Wang Y, et al. A novel liposome adjuvant DPC mediates Mycobacterium tuberculosis
subunit vaccine well to induce cell-mediated immunity and high protective efficacy in mice.
Vaccine. 2016 Mar 8;34(11):1370-8. doi: 10.1016/j.vaccine.2016.01.049. PubMed PMID: 26845736;
eng.
133. Guo LS. Liposome composition for treating dry eye. Google Patents; 1989.
134. Craig JP, Purslow C, Murphy PJ, et al. Effect of a liposomal spray on the pre-ocular tear film. Contact
Lens and Anterior Eye. 2010 2010/04/01/;33(2):83-87. doi:
https://doi.org/10.1016/j.clae.2009.12.007.

t
135. Morand K, Bartoletti AC, Bochot A, et al. Liposomal amphotericin B eye drops to treat fungal

ip
keratitis: Physico-chemical and formulation stability. International Journal of Pharmaceutics. 2007
2007/11/01/;344(1):150-153. doi: https://doi.org/10.1016/j.ijpharm.2007.04.028.

cr
136. Taha EI, El-Anazi MH, El-Bagory IM, et al. Design of liposomal colloidal systems for ocular delivery of
ciprofloxacin. Saudi Pharmaceutical Journal. 2014 2014/07/01/;22(3):231-239. doi:
https://doi.org/10.1016/j.jsps.2013.07.003.

us
137. Huang Y, Tao Q, Hou D, et al. A novel ion-exchange carrier based upon liposome-encapsulated
montmorillonite for ophthalmic delivery of betaxolol hydrochloride. International Journal of
Nanomedicine. 2017 03/02;12:1731-1745. doi: 10.2147/IJN.S122747. PubMed PMID: PMC5340245.
an
138. Manca ML, Matricardi P, Cencetti C, et al. Combination of argan oil and phospholipids for the
development of an effective liposome-like formulation able to improve skin hydration and allantoin
dermal delivery. Int J Pharm. 2016 May 30;505(1-2):204-11. doi: 10.1016/j.ijpharm.2016.04.008.
M
PubMed PMID: 27063848; eng.
139. Li X, Wang C, Xiao J, et al. Fibroblast growth factors, old kids on the new block. Seminars in cell &
developmental biology. 2016 01/06;53:155-167. doi: 10.1016/j.semcdb.2015.12.014. PubMed
d

PMID: PMC4875805.
140. Xu HL, Chen PP, ZhuGe DL, et al. Liposomes with Silk Fibroin Hydrogel Core to Stabilize bFGF and
e

Promote the Wound Healing of Mice with Deep Second-Degree Scald. Adv Healthc Mater. 2017
Oct;6(19). doi: 10.1002/adhm.201700344. PubMed PMID: 28661050; eng.
pt

141. Sahu K, Kaurav M, Pandey RS. Protease loaded permeation enhancer liposomes for treatment of
skin fibrosis arisen from second degree burn. Biomedicine & pharmacotherapy = Biomedecine &
ce

pharmacotherapie. 2017 Oct;94:747-757. doi: 10.1016/j.biopha.2017.07.141. PubMed PMID:


28800544; eng.
142. Azimi H, Fallah-Tafti M, Khakshur AA, et al. A review of phytotherapy of acne vulgaris: perspective
Ac

of new pharmacological treatments. Fitoterapia. 2012 Dec;83(8):1306-17. doi:


10.1016/j.fitote.2012.03.026. PubMed PMID: 22521501; eng.
143. Bergler-Czop B, Bilewicz-Stebel M, Stańkowska A, et al. Side effects of retinoid therapy on the
quality of vision. Acta Pharmaceutica2016. p. 471.
144. Rahman SA, Abdelmalak NS, Badawi A, et al. Tretinoin-loaded liposomal formulations: from lab to
comparative clinical study in acne patients. Drug Deliv. 2016 May;23(4):1184-93. doi:
10.3109/10717544.2015.1041578. PubMed PMID: 26004128; eng.
145. Jeong S, Lee J, Im BN, et al. Combined photodynamic and antibiotic therapy for skin disorder via
lipase-sensitive liposomes with enhanced antimicrobial performance. Biomaterials. 2017
Oct;141:243-250. doi: 10.1016/j.biomaterials.2017.07.009. PubMed PMID: 28697465; eng.
146. Doppalapudi S, Jain A, Chopra DK, et al. Psoralen loaded liposomal nanocarriers for improved skin
penetration and efficacy of topical PUVA in psoriasis. Eur J Pharm Sci. 2017 Jan 1;96:515-529. doi:
10.1016/j.ejps.2016.10.025. PubMed PMID: 27777066; eng.
147. Doppalapudi S, Mahira S, Khan W. Development and in vitro assessment of psoralen and
resveratrol co-loaded ultradeformable liposomes for the treatment of vitiligo. Journal of
photochemistry and photobiology B, Biology. 2017 Sep;174:44-57. doi:
10.1016/j.jphotobiol.2017.07.007. PubMed PMID: 28753523; eng.
148. Lee S-H, Sato Y, Hyodo M, et al. Size-Dependency of the Surface Ligand Density of Liposomes
Prepared by Post-insertion. Biological and Pharmaceutical Bulletin. 2017;40(7):1002-1009. doi:
10.1248/bpb.b16-00990.
149. Li H, Yuan D, Sun M, et al. Effect of ligand density and PEG modification on octreotide-targeted
liposome via somatostatin receptor in vitro and in vivo. Drug delivery. 2016

t
2016/11/21;23(9):3562-3572. doi: 10.1080/10717544.2016.1209797.

ip
150. Chu C, Xu P, Zhao H, et al. Effect of surface ligand density on cytotoxicity and pharmacokinetic
profile of docetaxel loaded liposomes. Asian Journal of Pharmaceutical Sciences. 2016

cr
2016/10/01/;11(5):655-661. doi: https://doi.org/10.1016/j.ajps.2016.04.001.
151. Toh M-R, Chiu GNC. Liposomes as sterile preparations and limitations of sterilisation techniques in
liposomal manufacturing. Asian Journal of Pharmaceutical Sciences. 2013 2013/04/01/;8(2):88-95.

us
doi: https://doi.org/10.1016/j.ajps.2013.07.011.
152. Singh B, Mundlamuri R, Friese T, et al. Benchmarking of Sterilizing-Grade Filter Membranes with
Liposome Filtration. PDA journal of pharmaceutical science and technology. 2018;72(3):223-235.
an
153. Solomon D, Gupta N, Mulla NS, et al. Role of In Vitro Release Methods in Liposomal Formulation
Development: Challenges and Regulatory Perspective. 2017 Nov;19(6):1669-1681. doi:
10.1208/s12248-017-0142-0. PubMed PMID: 28924630.
M
154. Gaspar RS, Florindo HF, Silva LC, et al. Regulatory Aspects of Oncologicals: Nanosystems Main
Challenges. In: Alonso MJ, Garcia-Fuentes M, editors. Nano-Oncologicals: New Targeting and
Delivery Approaches. Cham: Springer International Publishing; 2014. p. 425-452.
d

155. Sainz V, Conniot J, Matos A, et al. Regulatory Aspects on Nanomedicines. 2015.


e
pt
ce
Ac
Graphical abstract

t
ip
cr
us
an
M
e d
pt

The figure showing different application of liposome as a drug delivery system for example in skin disorders
ce

(skin cancer, burns and infection), ocular disorders (conjunctivitis and glaucoma), cancer therapy (ovarian
cancer), and vaccine delivery (tuberculosis).
Ac
Table1. liposomal formulation recently used in the global liposomes drug delivery market and some others still under

clinical trials.

Active drug Brand name Route of Lipid composition Indication Company


administration
Bupivacaine Exparel® Infiltration, Cholesterol, Interscalene brachial plexus Pacira
interscalene DPPG, and DEPC nerve block to produce post- Pharmaceuticals,
Branchial surgical regional analgesia Inc.
Plexus Nerve following shoulder surgery
Block in adults
Zoster Vaccine SHINGRIX® Intramuscular DOPC and Prevention of herpes zoster GlaxoSmithKline
Recombinant, cholesterol (shingles) in adults aged 50 Biologicals

t
ip
Adjuvanted years and older
Irinotecan Onivyde® Intravenous DSPC and Pancreatic Cancer Merrimack
MPEG-2000- Pharmaceuticals,

cr
DSPE Inc.
Doxorubicin DOXIL® Intravenous Cholesterol, Treatment of ovarian Janssen Products,
hydrochloride HSPC and cancer, AIDS-related L.P.

us
MPEG-DSPE. Kaposi's sarcoma, and
multiple myeloma
Vincristine sulfate Marqibo® Intravenous Sphingomyelin
an Acute lymphoblastic Spectrum
and cholesterol leukemia Pharmaceuticals,
Inc.
Cytarabine and Vyxeos® Intravenous DSPC, DSPG, Acute myeloid leukemia Jazz
M
daunorubicin and cholesterol Pharmaceuticals plc
Cytarabine DepoCyt® Intrathecal Cholesterol, Intrathecal treatment of Pacira
administration DOPC, and lymphomatous meningitis Pharmaceuticals,
d

DPPG. Inc.
Marqibo®, Phase 1 Intravenous Sphingomyelin Relapsed acute Spectrum
e

dexamethasone, and cholesterol lymphoblastic leukemia Pharmaceuticals, Inc


mitoxantrone, and (ALL)
pt

asparaginase (UK
ALL R3)
Pomalidomide in Phase 1 Intravenous Cholesterol, Pomalidomide in National Cancer
ce

combination with HSPC and combination with Institute (NCI)


doxorubicin MPEG-DSPE. doxorubicin liposome in
liposome people with advanced
Ac

refractory Kaposi sarcoma


Daunorubicin- Phase 1/2 Intravenous Cholesterol, Study of CPX-351 alone Children's Oncology
cytarabine (CPX- DSPC, and DSPG followed by fludarabine, Group and National
351) cytarabine, and filgrastim Cancer Institute
for children with relapsed (NCI)
acute myeloid leukemia
(AML)
Cisplatin Phase 1/2 Intravenous MPEG-DSPE and Evaluate the safety and Oncology Venture
egg PC tolerability of LiPlaCis
(liposomal cisplatin
formulation) in patients with
advanced or refractory
tumors
Cytarabine, Phase 2 Intravenous Cholesterol, Study of the impact of National Cancer
idarubicin, DSPC, and DSPG clinicogenetic risk-stratified Institute (NCI),
decitabine, and management on outcomes University of
liposome- of acute myeloid leukemia Nebraska
encapsulated in older patients
daunorubicin-
cytarabine
farletuzumab Phase 2 Intravenous Cholesterol, Assess the efficacy and Sponsor:
(MORAb 003) in HSPC and safety of farletuzumab Morphotek
combination with MPEG-DSPE. (MORAb 003) in Collaborator:
carboplatin plus combination with Eisai Co., Ltd
paclitaxel or carboplatin plus paclitaxel

t
carboplatin plus or carboplatin plus

ip
pegylated liposomal pegylated liposomal
doxorubicin (PLD) doxorubicin (PLD) in

cr
subjects with low CA125
platinum-sensitive ovarian
cancer

us
Vincristine Sulfate Phase 2 Intravenous Sphingomyelin Relapsed or refractory acute Wake Forest
and cholesterol myeloid leukemia (AML) University Health
an Sciences
pegylated liposomal Phase 2 Intravenous Cholesterol, Combination therapy for M.D. Anderson
doxorubicin, HSPC and patients with localized Cancer Center
bevacizumab, and MPEG-DSPE. triple-negative breast cancer
M
everolimus (TNBC) with tumors
predicted insensitive to
standard neoadjuvant
chemotherapy
d

Cytarabine Phase 2 Intrathecal Cholesterol, Cytarabine liposome for University of


DOPC, and adults with acute California, San
e

DPPG lymphoblastic leukemia or Diego


pt

acute lymphoblastic
lymphoma
doxorubicin Phase 2 Intravenous Cholesterol, Treating patients National Cancer
ce

hydrochloride, MPEG-DSPE, with stage II-III breast Institute (NCI),


epirubicin HSPC cancer that does not have Rutgers Cancer
hydrochloride, estrogen receptors, Institute of New
Ac

carboplatin, and progesterone receptors, or Jersey


paclitaxel large amounts of
(HER2)/neu protein (triple
negative).
Pegylated liposomal Phase 2/3 Intravenous Cholesterol, treating patients with National Cancer
doxorubicin HSPC and recurrent ovarian, fallopian Institute (NCI)
hydrochloride with MPEG-DSPE. tube, or primary peritoneal
atezolizumab and/or cancer
bevacizumab
Irinotecan Phase 2/3 Intravenous DSPC and Study of Irinotecan injection Ipsen Medical
MPEG-2000- (ONIVYDE®) versus Director
DSPE Topotecan in Patients with
Small Cell Lung Cancer
Doxorubicin Phase 3 Intravenous DPPC, MSPC and Study of ThermoDox® in Celsion Corporation
(ThermoDox®) DSPE-PEG2000 hepatocellular carcinoma (NASDAQ:CLSN)
(HCC) Using standardized
radiofrequency ablation
(RFA)
Vincristine Phase 3 Intravenous Sphingomyelin Improvement of outcome Spectrum
liposome, Rituximab and cholesterol and reduction of toxicity in Pharmaceuticals, Inc
elderly patients with CD20+ University Hospital,
aggressive B-cell Saarland
lymphoma

DPPG, 1,2-dipalmitoyl-sn-glycero-3-phospho-(1-racglycerol) (sodium salt); DEPC, 1,2-dierucoyl phosphatidylcholine;

t
DOPC, dioleoyl phosphatidylcholine; DSPC, 1,2-distearoyl-sn-glycero-3-phosphocholine; MPEG-2000-DSPE,

ip
Cholesterol N-(carbonyl-methoxy polyethylene glycol-2000)-1, 2-distearoly-sn-glycero-3-
phosphoethanolamine; HSPC, hydrogenated soy phosphatidylcholine; DSPG, distearoyl

cr
phosphatidylglycerol; egg PC, egg yolk phosphatidylcholine; HER2, human epidermal growth factor
receptor 2.

us
an
M
e d
pt
ce
Ac
Table 2. Liposome for gene and anticancer drugs delivery

Anticancer Lipid component/ Method of


Application Reference
drugs/gene polymers preparation
siRNA DOPC, DSPE-PEG, Ethanol injection Enhancing cellular association and gene [1]
STR-R8 silencing capacity exhibited by R8-PLPs
Anti-VEGF DSPC, PEG-DSPE, Thin layer Cellular delivery of anti-VEGF siRNA in [2]
siRNA DOPE hydration method SKBR-3 breast tumor cell line
(bevasiranib)
siRNA PEG-DSPE, Thin-film Enhancing cellular uptake of siRNA and gene [3]
DOTAP, NIPAAm, hydration method silencing activity by using temperature-
DMAPAAm responsive liposome
Paclitaxel and LHSSG2C14, Film dispersion Overcoming PTX resistance and improving the [4]
anti-survivin LHG2C14 method antitumor effect of PTX and down-regulation of

t
siRNA survivin overexpression

ip
Anti-miR-191 Lipoid S 100, stearyl Thin-film Efficient delivery of anti-miR-191 in SA [5]
amine (SA) hydration method liposome complex for inhibition of breast cancer
cells

cr
Plasmid DNA EPC, DOPE, Thin film hydration Formation of pDNA/protein /liposome [6]
(pVAX1GFP) DOTAP method complexes for more efficient pDNA delivery

us
pDNA and DOPE, DPPC, Poly Thin film hydration Using PEI based, liposome for Improvement [7]
siRNA ethylenimine method transfection efficiency and low cytotoxicity
siRNA DPPC, DPPG, Thin film hydration Reverse doxorubicin resistance of [8]
(silencing DPTAP, PDADMAC method OVCAR8/ADR cells (ovarian cancer).
an
ABCB1 gene),
Doxorubicin
Antimycin A DOPE, cholesterol Thin film Enhancement of the effective delivery and [9]
dehydration and stability of the incorporated drug to the
M
rehydration method mitochondria of A549 cells.
pDNA DOPE, OH-Chol Thin film hydration Explaining the size-dependent endocytosis [10]
method, pathway and the intracellular trafficking of
ultrasonication cationic lipoplexes using bone marrow-derived
d

methods dendritic cells (BMDCs).


Poly(L-lysine)- PC: DSPE-PEG, Thin film hydration Increasing gene silencing efficiency and [11]
e

siRNA complex EpCAM antibody method targeting specificity in breast cancer.


PAC1Rantisense DOPC, DOTAP, Thin film hydration Using light-responsive delivery strategy to [12]
pt

oligonucleotides Verteporfin method achieve enhanced endosome and lysosome


escape and PAC1Rgene silencing in PC12 cells
HIF-1α siRNA Soybean lecithin Thin film hydration siRNA-fc-LPs reduce the production of HIF-1α- [13]
ce

(S100), Folic acid, method associated protein and induce the apoptosis of
oleic acid, hypoxia-tolerant melanoma cells.
cholesterol, chitosan
Ac

Anti-MUC4 DPPC, DSPE, PEG Thin film hydration Higher targeting affinity and improving [14]
antibodies, method antiproliferative effect in PDAC
Gemcitabine
Gemcitabine DOPE, EPC Freezing/thawing Significant enhancement of the cytotoxicity, [15]
method anti-migration, and anti-colony formation
abilities of GEM through targeting of CD44
expressed on BCSCs by HA-conjugated
liposomes
Methotrexate PC, PI Thin film MTX-DG liposomes showing higher plasma [16]
diglyceride ester hydration, concentration, lower toxicity and retarded
freezing/thawing lymphoma growth rate as compared with MTX
method in mice bearing T-cell leukemic lymphoma.
Methotrexate PC, CH, PEG2000‐ Reverse‐phase Reduction the proliferation of human [17]
PE evaporation, thin lymphoblastic cell line K562 and significant
film hydration inhibition of RNA synthesis
method
Carfilzomib and Methoxy PEG2000- Thin film hydration Explaining Synergistic effect dual drug loaded [18]
Doxorubicin DSPE, DSPC, method liposomes in vitro and their efficacy in inhibiting
DPPE- glutaryl tumor growth in vivo reducing systemic toxicity
on multiple myeloma tumor cells.
Pemetrexed DOPC, DOPE, Reverse-phase Evaluation of liposomal membrane fluidity and [19]
HSPC, POPC, PEG evaporation its effect on drug release inside the murine
mesothelioma-xenograft model
Doxorubicin DSPC, DSPE-PEG Ethanol injection Stealth PoP liposomes provide long circulation [20]
method half-life and higher stability in storage for
months for the anticancer drug
Doxorubicin EPC, DOPE, PEG, Rotary evaporation Dual-targeted liposomes represent an effective [21]
Rhodamine folic followed by freeze cytotoxic formulation and higher tumor growth

t
acid, transferrin drying inhibition in human cervical carcinoma and

ip
A2780-ADR ovarian carcinoma cell monolayers

cr
siRNA, short interfering RNA; DOPC, 1,2-dioleoyl-sn-glycero-3-phosphocholine; DSPE-PEG,
1,2-distearoyl-sn-glycero-3-phosphoethanolamine- N-[methoxy(polyethylene glycol)-2000];

us
STR-R8, stearylated octaarginine; DSPC, 1,2-distearoyl-sn-glycero-3-phosphocholine; DOPE,
1,2-dioleoyl-sn-glycero-3- phosphoethanolamine; NIPAAm , 2.1.1 N-isopropylacrylamide;
DMAPAAm, N,N’-dimethylami- nopropylacrylamide; DOTAP, N-[1-(2,3-

double-stranded RNA virus;


an
dioleoyloxy)propyl]-N,N, N-trimethylammonium methyl-sulfate; Reovirus, a non-enveloped
LHSSG2C14, ditetradecyl 2- (4-(2-(2-(2-(2-(2,6-
diaminohexanamido)-3-(1H-imidazol-4-yl) propanamido) ethyl) disulfanyl) ethyl amino)-4-
M
oxobutanamido) pentanedioate; LHG2C14, ditetradecyl 2-(2-(2,6-diaminohexanamido)-3-(1H-
imidazol-4-yl) propan-amido) pentanedioate; Anti-miR-191(microRNA); DPPC,
dipalmitoylphosphatidylcholine; DPPG, 1,2-dipalmitoyl-sn-glycero-3-phospho-(1-racglycerol)
(sodium salt); DPTAP, 1,2-stearoyl-3-trimethylammonium-propane(chloride salt);
d

PDADMAC, poly(diallyldimethylammonium chloride); OH-Chol, cholesteryl-3β-


e

carboxyamido ethylene-N-hydroxyethylamine; EpCAM, epithelial cell adhesion molecule;


HIF-1α, hypoxia-inducible factor-1α; siRNA, small interfering RNA; PDAC, pancreatic
pt

ductal adenocarcinoma; HSPC, hydrogenated soy phosphatidylcholine; POPC, palmitoyloleoyl


phosphatidylcholine; BCSCs, breast cancer stem cells; PI, phosphatidylinositol; LPC,
lysophosphatidylcholine; LPE, lysophosphatidylethanolamine; LPG, lysophosphatidylglycerol.
ce
Ac
Table 3. Different substances incorporated into liposomes used for generation and potentiation of the immune
response

API Material Disease/purpose Route of Application References


used in the administration
liposome
composition
H5N1 vaccine JVRS-100 H5N1 Intramuscular Addition of JVRS-100 to H5N1 vaccine- [22]
(pDNA with enhanced immunogenicity and cross-
DOTIM/ protection against lethal H5N1 virus disease
cholesterol) in ferrets
SLP and DOTAP and Antigen delivery Intradermal Liposome enhances the delivery of the SLP [23]
poly(I:C) DOPC inducing an and to DCs in vitro and induced a functional

t
immune subcutaneous CD8+ T cell immune response in vivo to the

ip
response immunization CTL epitope present in the SLP.
P5 peptide and DOTAP and Murine tumor Subcutaneous Formulation enhance the release of [24]
Poly (I: C). cholesterol Interferon (IFN)-γ and induce strong

cr
antitumor responses
SP-Ld with FA Lecithin and Visceral Subcutaneous Liposomal formulation induces a long- [25]
cholesterol leishmaniasis lasting protective immune response in mice

us
and in hamsters against a challenge from
virulent L. donovani.
(IRV) HSPC and Rabies Intraperitoneal Liposomal rabies vaccine showed higher [26]
cholesterol injection
an survival rate and enhancement of the
immune response of mice to rabies vaccine
(Higher levels of
interleukin-2 (p < 0.05), interferon-c (p <
0.01), and natural killer cell activity (p <
M
0.05))
Lipopeptide- DDAB, Group A Intranasal Lipopeptides entrapped by liposomes induce [27]
based vaccine DPPC Streptococcus both mucosal and systemic immunity (higher
d

candidates IgG and IgA antibody titres) and also


against GAS produce more balanced and long lasting
e

Th1/Th2 responses in mice


Class B CpG DOTAP: Infectious Intranasal Cationic liposome harboring CpG ODN [28]
pt

ODNs DC- diseases induces both antigen-specific mucosal IgA


cholesterol responses and balanced Th1/Th2 responses.
Anti-Nicotine DOTAP, Nicotine Subcutaneous NsL NPs based anti-nicotine vaccine induce [29]
ce

Vaccine ( DSPE- PEG dependence high titer and high affinity of nicotine-
O-succinyl- 2000 specific antibodies in mice providing a
3hydroxmethyl- promising candidate in treating nicotine
(±) nicotine ) dependence
Ac

CRX-601 DOPC, PE- Influenza Sublingual The coating of modified liposomes produced [30]
PEG2K the most effective specific sublingual
immune response against influenza
Hepatitis B DPPC, Hepatitis B virus Transcutaneous (DMA)-based TCI system loaded with [31]
DNA vaccine cholesterol, immunization cationic liposomes help in improving the
DDA via immunogenicity of DNA vaccine.
microneedles
F1-V DOTAP, Plague Intranasal Liposome-polymer hybrid NPs stimulates [32]
DOPE, H A, stronger humoral and cellular immune
PEG responses. And provide Intranasal
vaccination against Yersinia pestis
GPC3-derived DOPE, Hepatocellular Intradermal Vaccination with pGPC3-liposome inhibited [33]
CTL epitope DOPG, carcinoma GPC3-expressing tumor growth in
peptide DOPC, hepatocellular carcinoma
cholesterol
AMM DDA, Poly Mycobacterium Subcutaneous AMM Liposomal formulation has a good [34]
I:C, tuberculosis stability and induces Th1-type cell-mediated
cholesterol immune responses. and also provide long-
(DPC) term protection against mice M. tuberculosis
infection
Gp2 DMPC, Breast cancer Subcutaneous Liposomal formulation showed the highest [35]
DMPG, number of IFN-γ+ in CD8+ cells and
DOPE, cytotoxic T lymphocyte response.
cholesterol The immunization led to lower tumor sizes
and longer survival time against a breast
cancer model overexpressing
HER2/neu.

t
S19-OMP DOPE, Brucella abortus Subcutaneous (S19-OMP-liposome) showed enhanced [36]

ip
DODAP infection protection compared to groups of mice
inoculated with S19 OMP alone and S19 live
B. abortus vaccine with higher immune

cr
response (Th1 based cellular immunity)
PCV-2 Soybean Porcine Subcutaneous RGPL showed excellent particle stability [37]

us
attenuated phospholipid, circovirus type 2 with a strong IgG response and increase the
antigen cholesterol, infection production of Th1 and Th2 associated IgG
RGPL subtypes and cytokines
an
Poly (I:C), polyriboinosinic: polyribocytidylic acid; DOTAP, dioleoyl-3-trimethylammonium propane;
M
SP-Ld, L.donovani intracellular serine protease; FA, Freund’s adjuvant; IRV, inactivated rabies
vaccine; DDAB, dimethyldiocta decyl ammonium bromide; GAS, group A Streptococcus; ODNs,
oligodeoxynucleotides; CpG, containing immunostimulatory motifs; NsL NPs, nanohorn supported
d

liposome nanoparticles; PE-PEG2K, [N-(carbonyl-methoxy polyethyleneglycol-2000)-1,2-distearoyl-


e

sn-glycero-3-phosphoethanol amine sodium salt; DDA, dimethyldioctadecylammonium; DMA,


pt

dissolving microneedle array; TCI, transcutaneous immunization; F1-V, a candidate recombinant


antigen for Yersinia pestis; H A, hyaluronic acid; HCC, hepatocellular carcinoma; GPC3, an oncofetal
ce

antigen overexpressed in HCC; AMM, subunit vaccine Ag85B-MPT64(190-198)-Mtb8.4; DDA,


dioctade-cylammonium; Gp2, a HER2/neu-derived peptide; DMPC, dimyristoylphosphatidylcoline;
DMPG, dimyristoylphosphoglycerol; OMP, outer membrane protein of Brucella abortus S19 vaccine;
Ac

DODAP, dioleoyl-3-dimethylammonium-propane; RGPL, R. glutinosa polysaccharide.


Table 4. Substances encapsulated into liposomes for wound healing

API Liposomal Application References


composition
Danggui Soybean DBLTG formulation was successfully leading to faster wound [38]
Buxue Extract phospholipid and closure, higher hydroxyl proline levels, and provide topical
cholesterol sustained release drug delivery systems.
bFGF Phospholipids, silk SF-bFGF-LIP formulation enhances bFGF stability (More than 50% [39]
fibroin of free bFGF are degraded after 8 h of incubation, while only 18.6%
of the encapsulated bFGF in SF-LIP are destroyed), accelerates the
wound closure of mice with deep second-degree scald.
Gatifloxacin Phospholipon 90 Lyophilized liposomal wafers provided localization and sustained [40]
(GTX) H, cholesterol release of the drug (GTX gel without liposomes 54.53% released
while from the liposomal gel was 33.04% after 12 hrs.) to the

t
wound site.

ip
Hemoglobin PEG, inositol LEH enhances surface blood flow and accelerates skin wound [41]
hexaphosphate, healing in diabetic mice
2,3-

cr
diphosphoglycerate
GHK-Cu DOPC, DOPG GHK-Cu-liposomes enhance the expression of both growth factors [42]

us
(VEGF and FGF-2), and decrease the wound healing time in
comparison with the free GHK-Cu, In a mice scald model
Povidone– Phospholipids Liposomal 3% PVP–I hydrogel provide better scores in subjective [43]
Iodine assessments of quality of wound healing, and can be used as a
an
dressing for non-infected MSGs
Quercetin PC and cholesterol Liposomal formulations produce sustained release of drug in wound [44]
areas (in vitro)
siRNA DOTAP LPP formulation enhances delivery of siRNA targeted against [45]
M
Keap1 and accelerates diabetic tissue regeneration in humanized
Murine diabetic wound.
Cysteine PC, propylene PG-liposomes enhance the deposition of papain and the skin [46]
protease glycol (PG) permeation efficiency in thermal burn-induced wound
d

(papain) Fibrosis.
Madecassoside Egg yolk lecithin, MA double-emulsion liposomes had better stability and more [47]
e

(MA) cholesterol excellent abilities of permeation and distribution in the burned skin
with superior performance in wound healing.
pt

Usnic acid PC, gelatin UAL provide greater collagen. Deposition, development, and [48]
maturation of granulation tissue and scar repair in the porcine
model
ce

DBLTG, Danggui Buxue extract-loaded liposomes in the thermosensitive gel; bFGF, fibroblast
growth factor; LEH, liposome-encapsulated hemoglobin; (GHK)-Cu, glycyl-l-histidyl-l-lysine;
MSGs, meshed skin grafts; LPP, lipoproteoplex.
Ac
References

1. Fisher RK, Mattern-Schain SI, Best MD, et al. Improving the efficacy of liposome-mediated vascular
gene therapy via lipid surface modifications. Journal of Surgical Research. 2017;219:136-144.
2. Golkar N, Samani SM, Tamaddon AM. Modulated cellular delivery of anti-VEGF siRNA (bevasiranib)
by incorporating supramolecular assemblies of hydrophobically modified polyamidoamine
dendrimer in stealth liposomes. International journal of pharmaceutics. 2016;510(1):30-41.
3. Wang J, Ayano E, Maitani Y, et al. Enhanced cellular uptake and gene silencing activity of siRNA
using temperature-responsive polymer-modified liposome. International journal of pharmaceutics.
2017;523(1):217-228.
4. Chen X, Zhang Y, Tang C, et al. Co-delivery of paclitaxel and anti-survivin siRNA via redox-sensitive
oligopeptide liposomes for the synergistic treatment of breast cancer and metastasis. International

t
Journal of Pharmaceutics. 2017;529(1-2):102-115.

ip
5. Sharma S, Rajendran V, Kulshreshtha R, et al. Enhanced efficacy of anti-miR-191 delivery through
stearylamine liposome formulation for the treatment of breast cancer cells. International Journal of

cr
Pharmaceutics. 2017;530(1-2):387-400.
6. Alves RF, Favaro MT, Balbino TA, et al. Recombinant protein-based nanocarriers and their
association with cationic liposomes: Characterization and in vitro evaluation. Colloids and Surfaces

us
A: Physicochemical and Engineering Aspects. 2017;513:1-10.
7. Pinnapireddy SR, Duse L, Strehlow B, et al. Composite liposome-PEI/nucleic acid lipopolyplexes for
safe and efficient gene delivery and gene knockdown. Colloids and Surfaces B: Biointerfaces.
an
2017;158:93-101.
8. Xia Y, Wang X, Cheng H, et al. A polycation coated liposome as efficient siRNA carrier to overcome
multidrug resistance. Colloids and Surfaces B: Biointerfaces. 2017;159:427-436.
M
9. Mallick S, Lee S, Park J-I, et al. Liposomes containing Cholesterol and Mitochondria-Penetrating
Peptide (MPP) for targeted delivery of Antimycin A to A549 cells. Colloids and Surfaces B:
Biointerfaces. 2017.
10. Inoh Y, Nagai M, Matsushita K, et al. Gene transfection efficiency into dendritic cells is influenced
d

by the size of cationic liposomes/DNA complexes. European Journal of Pharmaceutical Sciences.


e

2017;102:230-236.
11. Bhavsar D, Subramanian K, Sethuraman S, et al. ‘Nano–in–nano’hybrid liposomes increase target
pt

specificity and gene silencing efficiency in breast cancer induced SCID mice. European Journal of
Pharmaceutics and Biopharmaceutics. 2017;119:96-106.
ce

12. Chen W, Deng W, Goldys EM. Light-Triggerable Liposomes for Enhanced Endolysosomal Escape and
Gene Silencing in PC12 Cells. Molecular Therapy-Nucleic Acids. 2017;7:366-377.
13. Chen Z, Zhang T, Wu B, et al. Insights into the therapeutic potential of hypoxia-inducible factor-1α
small interfering RNA in malignant melanoma delivered via folate-decorated cationic liposomes.
Ac

International journal of nanomedicine. 2016;11:991.


14. UREY C, HILMERSSON KS, ANDERSSON B, et al. Development and In Vitro Characterization of a
Gemcitabine-loaded MUC4-targeted Immunoliposome Against Pancreatic Ductal Adenocarcinoma.
Anticancer Research. 2017;37(11):6031-6039.
15. Han N-K, Shin DH, Kim JS, et al. Hyaluronan-conjugated liposomes encapsulating gemcitabine for
breast cancer stem cells. International journal of nanomedicine. 2016;11:1413.
16. Alekseeva AA, Moiseeva EV, Onishchenko NR, et al. liposomal formulation of a methotrexate
lipophilic prodrug: assessment in tumor cells and mouse T-cell leukemic lymphoma. International
Journal of Nanomedicine. 2017;12:3735.
17. Ciobanu AM, Bârcă M, Manda G, et al. Methotrexate Liposomes-A Reliable Therapeutic Option.
Liposomes: InTech; 2017.
18. Ashley JD, Quinlan CJ, Schroeder VA, et al. Dual Carfilzomib and Doxorubicin–Loaded Liposomal
Nanoparticles for Synergistic Efficacy in Multiple Myeloma. Molecular cancer therapeutics.
2016;15(7):1452-1459.
19. Eldin NE, Lila ASA, Kawazoe K, et al. Encapsulation in a rapid-release liposomal formulation
enhances the anti-tumor efficacy of pemetrexed in a murine solid mesothelioma-xenograft model.
European Journal of Pharmaceutical Sciences. 2016;81:60-66.
20. Luo D, Carter KA, Razi A, et al. Doxorubicin encapsulated in stealth liposomes conferred with light-
triggered drug release. Biomaterials. 2016;75:193-202.
21. Sriraman SK, Salzano G, Sarisozen C, et al. Anti-cancer activity of doxorubicin-loaded liposomes co-
modified with transferrin and folic acid. European journal of pharmaceutics and biopharmaceutics :
official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik eV. 2016
Aug;105:40-9. doi: 10.1016/j.ejpb.2016.05.023. PubMed PMID: 27264717; PubMed Central PMCID:

t
PMCPmc4931959. eng.

ip
22. Liu F, Sun X, Fairman J, et al. A cationic liposome-DNA complexes adjuvant (JVRS-100) enhances the
immunogenicity and cross-protective efficacy of pre-pandemic influenza A (H5N1) vaccine in

cr
ferrets. Virology. 2016 May;492:197-203. doi: 10.1016/j.virol.2016.02.024. PubMed PMID:
26967975; eng.
23. Varypataki EM, van der Maaden K, Bouwstra J, et al. Cationic Liposomes Loaded with a Synthetic

us
Long Peptide and Poly(I:C): a Defined Adjuvanted Vaccine for Induction of Antigen-Specific T Cell
Cytotoxicity. The AAPS Journal. 2015 11/1206/23/received 10/09/accepted;17(1):216-226. doi:
10.1208/s12248-014-9686-4. PubMed PMID: PMC4287297.
an
24. Alipour Talesh G, Ebrahimi Z, Badiee A, et al. Poly (I:C)-DOTAP cationic nanoliposome containing
multi-epitope HER2-derived peptide promotes vaccine-elicited anti-tumor immunity in a murine
M
model. Immunology letters. 2016 Aug;176:57-64. doi: 10.1016/j.imlet.2016.05.016. PubMed PMID:
27260485; eng.
25. Das P, Paik D, Naskar K, et al. Leishmania donovani serine protease encapsulated in liposome elicits
protective immunity in experimental visceral leishmaniasis. Microbes and infection. 2017 Sep 29.
d

doi: 10.1016/j.micinf.2017.09.011. PubMed PMID: 28970116; eng.


e

26. Miao L, Yang Y, Yan M, et al. Enhanced Immune Response to Rabies Viruses by the Use of a
Liposome Adjuvant in Vaccines. Viral immunology. 2017 Sep 27. doi: 10.1089/vim.2017.0093.
pt

PubMed PMID: 28953450; eng.


27. Ghaffar KA, Marasini N, Giddam AK, et al. Liposome-based intranasal delivery of lipopeptide
ce

vaccine candidates against group A streptococcus. Acta biomaterialia. 2016 Sep 01;41:161-8. doi:
10.1016/j.actbio.2016.04.012. PubMed PMID: 27063491; eng.
28. Tada R, Muto S, Iwata T, et al. Attachment of class B CpG ODN onto DOTAP/DC-chol liposome in
nasal vaccine formulations augments antigen-specific immune responses in mice. BMC research
Ac

notes. 2017;10(1):68.
29. Zheng H, Hu Y, Huang W, et al. Negatively charged carbon nanohorn supported cationic liposome
nanoparticles: a novel delivery vehicle for anti-nicotine vaccine. Journal of biomedical
nanotechnology. 2015;11(12):2197-2210.
30. Oberoi HS, Yorgensen YM, Morasse A, et al. PEG modified liposomes containing CRX-601 adjuvant
in combination with methylglycol chitosan enhance the murine sublingual immune response to
influenza vaccination. Journal of Controlled Release. 2016;223:64-74.
31. Qiu Y, Guo L, Zhang S, et al. DNA-based vaccination against hepatitis B virus using dissolving
microneedle arrays adjuvanted by cationic liposomes and CpG ODN. Drug delivery.
2016;23(7):2391-2398.
32. Fan Y, Sahdev P, Ochyl LJ, et al. Cationic liposome–hyaluronic acid hybrid nanoparticles for
intranasal vaccination with subunit antigens. Journal of controlled release. 2015;208:121-129.
33. Iwama T, Uchida T, Sawada Y, et al. Vaccination with liposome-coupled glypican-3-derived epitope
peptide stimulates cytotoxic T lymphocytes and inhibits GPC3-expressing tumor growth in mice.
Biochemical and biophysical research communications. 2016;469(1):138-143.
34. Liu X, Da Z, Wang Y, et al. A novel liposome adjuvant DPC mediates Mycobacterium tuberculosis
subunit vaccine well to induce cell-mediated immunity and high protective efficacy in mice.
Vaccine. 2016 Mar 8;34(11):1370-8. doi: 10.1016/j.vaccine.2016.01.049. PubMed PMID: 26845736;
eng.
35. Razazan A, Behravan J, Arab A, et al. Conjugated nanoliposome with the HER2/neu-derived peptide
GP2 as an effective vaccine against breast cancer in mice xenograft model. PloS one.
2017;12(10):e0185099. doi: 10.1371/journal.pone.0185099. PubMed PMID: 29045460; PubMed

t
Central PMCID: PMCPmc5646774. eng.

ip
36. Mukherjee F, Prasad A, Bahekar V, et al. Evaluation of immunogenicity and protective efficacy of a
liposome containing Brucella abortus S19 outer membrane protein in BALB/c mice. Iranian journal

cr
of veterinary research. 2016;17(1):1.
37. Huang Y, Liu Z, Bo R, et al. The enhanced immune response of PCV-2 vaccine using Rehmannia
glutinosa polysaccharide liposome as an adjuvant. International journal of biological

us
macromolecules. 2016;86:929-936.
38. Cui M-D, Pan Z-H, Pan L-Q. Danggui Buxue Extract-Loaded Liposomes in Thermosensitive Gel
Enhance In Vivo Dermal Wound Healing via Activation of the VEGF/PI3K/Akt and TGF-β/Smads
an
Signaling Pathway. Evidence-Based Complementary and Alternative Medicine. 2017;2017.
39. Xu HL, Chen PP, ZhuGe DL, et al. Liposomes with Silk Fibroin Hydrogel Core to Stabilize bFGF and
Promote the Wound Healing of Mice with Deep Second‐Degree Scald. Advanced Healthcare
M
Materials. 2017;6(19).
40. Avachat AM, Takudage PJ. Design and Characterization of Multifaceted Lyophilized Liposomal
Wafers with Promising Wound Healing Potential. Journal of Liposome Research. 2017 (just-
d

accepted):1-52.
41. Fukui T, Kawaguchi AT, Takekoshi S, et al. Liposome‐Encapsulated Hemoglobin Accelerates Skin
e

Wound Healing in Diabetic dB/dB Mice. Artificial Organs. 2017;41(4):319-326.


42. Wang X, Liu B, Xu Q, et al. GHK‐Cu‐liposomes accelerate scald wound healing in mice by promoting
pt

cell proliferation and angiogenesis. Wound Repair and Regeneration. 2017;25(2):270-278.


43. Vogt PM, Hauser J, Mueller S, et al. Efficacy of Conventional and Liposomal Povidone–Iodine in
ce

Infected Mesh Skin Grafts: An Exploratory Study. Infectious Diseases and Therapy. 2017;6(4):545-
555.
44. Jangde R, Singh D. Preparation and optimization of quercetin-loaded liposomes for wound healing,
Ac

using response surface methodology. Artificial cells, nanomedicine, and biotechnology.


2016;44(2):635-641.
45. Rabbani PS, Zhou A, Borab ZM, et al. Novel lipoproteoplex delivers Keap1 siRNA based gene
therapy to accelerate diabetic wound healing. Biomaterials. 2017;132:1-15.
46. Sahu K, Kaurav M, Pandey RS. Protease loaded permeation enhancer liposomes for treatment of
skin fibrosis arisen from second degree burn. Biomedicine & Pharmacotherapy. 2017;94:747-757.
47. Li Z, Liu M, Wang H, et al. Increased cutaneous wound healing effect of biodegradable liposomes
containing madecassoside: preparation optimization, in vitro dermal permeation, and in vivo
bioevaluation. International journal of nanomedicine. 2016;11:2995.
48. Nunes PS, Rabelo AS, de Souza JCC, et al. Gelatin-based membrane containing usnic acid-loaded
liposome improves dermal burn healing in a porcine model. International journal of pharmaceutics.
2016;513(1):473-482.

You might also like